PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 2555360-12 1989 Secondary amine mono-oxygenase is unique in its ability to function as cytochrome P-450 in activating molecular oxygen but to do so with a myoglobin-like active site. Oxygen 21-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 2516815-0 1989 Metabolic activation of 7,12-dimethylbenz(a)anthracene: role of cytochrome P-450 isoenzymes in the formation of DNA and protein adducts in vitro. 7,12-dimethylbenz(a 24-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 2604729-1 1989 Effects of a cytochrome P-450 inhibitor on the formation and metabolism of oxygenated sterol products of lanosterol. Sterols 86-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 2604729-1 1989 Effects of a cytochrome P-450 inhibitor on the formation and metabolism of oxygenated sterol products of lanosterol. Lanosterol 105-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 2628054-0 1989 Species differences in specificity of hydrocarbon-inducible forms of cytochrome P-450. Hydrocarbons 38-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 2622477-3 1989 The present in vitro study was, therefore, designed to assess the influence of 5-fluorocytosine and 5-fluorouracil on the hepatic cytochrome P 450 concentration in human and rat liver microsomes. Flucytosine 79-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 2622477-3 1989 The present in vitro study was, therefore, designed to assess the influence of 5-fluorocytosine and 5-fluorouracil on the hepatic cytochrome P 450 concentration in human and rat liver microsomes. Fluorouracil 100-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 2597175-0 1989 Absence of an isotope effect in induction of cytochrome P-450 and xenobiotic metabolizing enzyme activities by stable isotope-labelled phenobarbital isotopomers. Phenobarbital 135-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 2509067-0 1989 Roles of individual human cytochrome P-450 enzymes in the bioactivation of benzo(a)pyrene, 7,8-dihydroxy-7,8-dihydrobenzo(a)pyrene, and other dihydrodiol derivatives of polycyclic aromatic hydrocarbons. Benzo(a)pyrene 75-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2509067-0 1989 Roles of individual human cytochrome P-450 enzymes in the bioactivation of benzo(a)pyrene, 7,8-dihydroxy-7,8-dihydrobenzo(a)pyrene, and other dihydrodiol derivatives of polycyclic aromatic hydrocarbons. benzo(a)pyrene 7,8-dihydrodiol 91-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2509067-0 1989 Roles of individual human cytochrome P-450 enzymes in the bioactivation of benzo(a)pyrene, 7,8-dihydroxy-7,8-dihydrobenzo(a)pyrene, and other dihydrodiol derivatives of polycyclic aromatic hydrocarbons. trans-1,2-dihydro-1,2-naphthalenediol 142-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2509067-0 1989 Roles of individual human cytochrome P-450 enzymes in the bioactivation of benzo(a)pyrene, 7,8-dihydroxy-7,8-dihydrobenzo(a)pyrene, and other dihydrodiol derivatives of polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 169-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2605239-0 1989 Isotopically labeled chlorobenzenes as probes for the mechanism of cytochrome P-450 catalyzed aromatic hydroxylation. Chlorobenzenes 21-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 2605239-1 1989 Noncompetitive and competitive intermolecular deuterium isotope effects were measured for the cytochrome P-450 catalyzed hydroxylation of a series of selectively deuterated chlorobenzenes. Deuterium 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 2605239-1 1989 Noncompetitive and competitive intermolecular deuterium isotope effects were measured for the cytochrome P-450 catalyzed hydroxylation of a series of selectively deuterated chlorobenzenes. deuterated chlorobenzenes 162-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 2605239-5 1989 These results eliminate initial epoxide formation and initial electron abstraction (charge transfer) as viable mechanisms for the cytochrome P-450 catalyzed hydroxylation of chlorobenzene. Epoxy Compounds 32-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 2605239-5 1989 These results eliminate initial epoxide formation and initial electron abstraction (charge transfer) as viable mechanisms for the cytochrome P-450 catalyzed hydroxylation of chlorobenzene. chlorobenzene 174-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 2504483-4 1989 Thiotepa was shown to be metabolized under these conditions in a NADPH- and O2-dependent reaction that was catalyzed by one or more microsomal cytochrome P-450 enzymes that were present in the S-9 fraction. Thiotepa 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 2688634-0 1989 Purification and characterization of an anticonvulsant-induced human cytochrome P-450 catalysing cyclosporin metabolism. Cyclosporine 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 2515662-2 1989 The anaerobic NADPH-reduction of the isozymes cytochrome P-450 LM2 and LM4 was used as a functional tool to study the component interaction in reconstituted monooxygenase systems in dependence on different phospholipids. NADP 14-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2515662-2 1989 The anaerobic NADPH-reduction of the isozymes cytochrome P-450 LM2 and LM4 was used as a functional tool to study the component interaction in reconstituted monooxygenase systems in dependence on different phospholipids. Phospholipids 206-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2573205-4 1989 As a result, famotidine should be considered the H2-receptor antagonist of choice for critically ill patients who require gastric-acid suppression and at the same time are being treated with other drugs that depend on the cytochrome P-450 mixed-function oxidase system for their metabolism and/or on renal tubular mechanisms for their excretion. Famotidine 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 2600595-0 1989 Demethylation of tertiary amines by a reconstituted cytochrome P-450 enzyme system: kinetics of oxygen consumption and hydrogen peroxide formation. tertiary amines 17-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 2600595-0 1989 Demethylation of tertiary amines by a reconstituted cytochrome P-450 enzyme system: kinetics of oxygen consumption and hydrogen peroxide formation. Oxygen 96-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 2600595-0 1989 Demethylation of tertiary amines by a reconstituted cytochrome P-450 enzyme system: kinetics of oxygen consumption and hydrogen peroxide formation. Hydrogen Peroxide 119-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 2600595-1 1989 Initial reaction rates of oxygen consumption and hydrogen peroxide formation in a cytochrome P-450 catalyzed reaction are practically independent of the nature of tertiary amines that were used as substrates. Oxygen 26-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 2600595-1 1989 Initial reaction rates of oxygen consumption and hydrogen peroxide formation in a cytochrome P-450 catalyzed reaction are practically independent of the nature of tertiary amines that were used as substrates. Hydrogen Peroxide 49-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 2600595-1 1989 Initial reaction rates of oxygen consumption and hydrogen peroxide formation in a cytochrome P-450 catalyzed reaction are practically independent of the nature of tertiary amines that were used as substrates. Amines 172-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 2600595-3 1989 Both hydrogen peroxide and water formation lower the efficiency of the monooxygenatic activity of cytochrome P-450. Hydrogen Peroxide 5-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 2600595-3 1989 Both hydrogen peroxide and water formation lower the efficiency of the monooxygenatic activity of cytochrome P-450. Water 27-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 2614264-0 1989 Mechanistic studies of lanosterol 14 alpha-methyl demethylase: substrate requirements for the component reactions catalyzed by a single cytochrome P-450 isozyme. Lanosterol 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-152 2614264-1 1989 Lanosterol 14 alpha-methyl demethylation is a cytochrome P-450-dependent process that proceeds through the oxidative sequence of alcohol, aldehyde followed by decarbonylation with formic acid release. Lanosterol 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2614264-1 1989 Lanosterol 14 alpha-methyl demethylation is a cytochrome P-450-dependent process that proceeds through the oxidative sequence of alcohol, aldehyde followed by decarbonylation with formic acid release. Alcohols 129-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2614264-1 1989 Lanosterol 14 alpha-methyl demethylation is a cytochrome P-450-dependent process that proceeds through the oxidative sequence of alcohol, aldehyde followed by decarbonylation with formic acid release. Aldehydes 138-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2614264-1 1989 Lanosterol 14 alpha-methyl demethylation is a cytochrome P-450-dependent process that proceeds through the oxidative sequence of alcohol, aldehyde followed by decarbonylation with formic acid release. formic acid 180-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2559440-0 1989 Oxidation of diethyldithiocarbamate to disulfiram by liver microsomal cytochrome P-450-containing monooxygenase system. Ditiocarb 13-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 2559440-0 1989 Oxidation of diethyldithiocarbamate to disulfiram by liver microsomal cytochrome P-450-containing monooxygenase system. Disulfiram 39-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 2559440-1 1989 We examined the involvement of cytochrome P-450 in the oxidation of diethyldithiocarbamate (DTC) to disulfiram (DS) by liver microsomes in the presence of NADPH. Ditiocarb 68-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 2559440-1 1989 We examined the involvement of cytochrome P-450 in the oxidation of diethyldithiocarbamate (DTC) to disulfiram (DS) by liver microsomes in the presence of NADPH. Ditiocarb 92-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 2559440-1 1989 We examined the involvement of cytochrome P-450 in the oxidation of diethyldithiocarbamate (DTC) to disulfiram (DS) by liver microsomes in the presence of NADPH. Disulfiram 100-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 2559440-1 1989 We examined the involvement of cytochrome P-450 in the oxidation of diethyldithiocarbamate (DTC) to disulfiram (DS) by liver microsomes in the presence of NADPH. Disulfiram 112-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 2559440-5 1989 With microsomes peroxidized by cumene hydroperoxide, the extent of NADPH-dependent DS production lowered in proportion to the decrease in cytochrome P-450. cumene hydroperoxide 31-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-154 2559440-5 1989 With microsomes peroxidized by cumene hydroperoxide, the extent of NADPH-dependent DS production lowered in proportion to the decrease in cytochrome P-450. NADP 67-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-154 2559440-6 1989 Inhibitors of cytochrome P-450 such as SKF-525A, metyrapone and n-octylamine dose-dependently inhibited the DS production. Proadifen 39-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 2559440-6 1989 Inhibitors of cytochrome P-450 such as SKF-525A, metyrapone and n-octylamine dose-dependently inhibited the DS production. Metyrapone 49-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 2559440-6 1989 Inhibitors of cytochrome P-450 such as SKF-525A, metyrapone and n-octylamine dose-dependently inhibited the DS production. octylamine 64-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 2559440-8 1989 It is concluded that oxidation of DTC to DS by liver microsomes largely proceeds via the cytochrome P-450-containing monooxygenase system and partly by hydrogen peroxide generated during NADPH oxidation. Ditiocarb 34-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 2559440-8 1989 It is concluded that oxidation of DTC to DS by liver microsomes largely proceeds via the cytochrome P-450-containing monooxygenase system and partly by hydrogen peroxide generated during NADPH oxidation. Disulfiram 41-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 2618083-4 1989 Spectral interactions of N-containing heteroaromatic compounds with the cytochrome P-450 system are type I or type II depending on the state of induction, and are relatively weak. Nitrogen 25-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 2550511-2 1989 This compound is an imidazole derivative, and therefore, its possible effect on cytochrome P-450-dependent enzyme activities in the adrenal gland was evaluated. imidazole 20-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 2504483-4 1989 Thiotepa was shown to be metabolized under these conditions in a NADPH- and O2-dependent reaction that was catalyzed by one or more microsomal cytochrome P-450 enzymes that were present in the S-9 fraction. NADP 65-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 2504483-4 1989 Thiotepa was shown to be metabolized under these conditions in a NADPH- and O2-dependent reaction that was catalyzed by one or more microsomal cytochrome P-450 enzymes that were present in the S-9 fraction. Oxygen 76-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 2504483-9 1989 These findings establish that the cytotoxic effects of thiotepa are oxygen dependent and may involve, at least in part, metabolic processes catalyzed by cytochrome P-450 enzymes. Thiotepa 55-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-169 2504483-9 1989 These findings establish that the cytotoxic effects of thiotepa are oxygen dependent and may involve, at least in part, metabolic processes catalyzed by cytochrome P-450 enzymes. Oxygen 68-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-169 2688863-8 1989 In contrast, N-demethylation of cocaine mediated by microsomal cytochrome P-450 produces norcocaine and this metabolite was shown to be pharmacologically active, the action being similar to cocaine. Cocaine 32-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 2775304-9 1989 The data in this paper are consistent with 4-hydroxylation of debrisoquine in both rat and human liver catalysed by a specific form of cytochrome P-450. Debrisoquin 62-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 2590684-1 1989 Crosslinking of protein molecules with bifunctional reagents and subsequent electrophoresis of the modified proteins revealed the presence of cytochrome P-450 LM 2 oligomers in proteoliposome membranes obtained in different ways and differing in their phospholipid composition. Phospholipids 252-264 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-158 2688863-8 1989 In contrast, N-demethylation of cocaine mediated by microsomal cytochrome P-450 produces norcocaine and this metabolite was shown to be pharmacologically active, the action being similar to cocaine. norcocaine 89-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 2690426-1 1989 Cytochrome P-450 appears to catalyse most monooxygenation reactions by sequential one-electron steps rather than by a single, concerted transfer of the ferryl oxygen to the substrate. Oxygen 46-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2572090-1 1989 Cimetidine, the first marketed histamine2-receptor antagonist, has been shown to decrease the clearance of warfarin consistently through inhibition of cytochrome P-450 metabolism. Cimetidine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 2747769-2 1989 Since cimetidine inhibits the cytochrome P-450-dependent biotransformation of numerous drugs, we investigated the possibility that it might also inhibit the cytochrome P-450--dependent metabolism of estradiol. Estradiol 199-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 2682943-3 1989 Rifampin and perhaps nafcillin induce cytochrome P-450-dependent isoenzyme metabolism of cyclosporine. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 2682943-3 1989 Rifampin and perhaps nafcillin induce cytochrome P-450-dependent isoenzyme metabolism of cyclosporine. Nafcillin 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 2682943-3 1989 Rifampin and perhaps nafcillin induce cytochrome P-450-dependent isoenzyme metabolism of cyclosporine. Cyclosporine 89-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 2747769-2 1989 Since cimetidine inhibits the cytochrome P-450-dependent biotransformation of numerous drugs, we investigated the possibility that it might also inhibit the cytochrome P-450--dependent metabolism of estradiol. Cimetidine 6-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 2590249-0 1989 [A study of the interaction of substrates with cytochrome P-450 by a method of UV- and 1H-NMR spectroscopy]. Hydrogen 87-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 2747769-2 1989 Since cimetidine inhibits the cytochrome P-450-dependent biotransformation of numerous drugs, we investigated the possibility that it might also inhibit the cytochrome P-450--dependent metabolism of estradiol. Cimetidine 6-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 2590249-1 1989 Acceleration of substrate longitudinal relaxation (T1) was used to study cytochrome P-450-aminopyrine (1st type substrate) and P-450-4-methoxypyridine (2nd type substrate) complexes. Aminopyrine 90-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 2803244-5 1989 The reasons for these differences have been investigated with particular reference to the mode of cytochrome P-450-catalysed decomposition of CHP, that is, via homolytic or heterolytic cleavage of the hydroperoxide dioxygen bond. hydroperoxide dioxygen 201-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 2803244-9 1989 It is suggested that the inability of CHP to support ECOD activity in the olfactory epithelium and the extensive inactivation of cytochrome P-450 that it causes both stem from decomposition of the hydroperoxide occurring homolytically rather than heterolytically in this tissue. Hydrogen Peroxide 197-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 2775621-5 1989 By contrast, ketoconazole had a much smaller effect on the 0-8 h urinary metabolic ratio of debrisoquine, indicating that ketoconazole has a selective inhibitory effect on different forms of cytochrome P-450. Ketoconazole 122-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-207 2778096-4 1989 This is most likely due to the saturation within therapeutic dosage range of the subspecies of cytochrome P-450 responsible for hydroxylation of DMT. desmethyltrimipramine 145-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 2474030-0 1989 Cytochrome P-450-dependent omega-oxidation of leukotriene B4 in rodent and human epidermis. Leukotriene B4 46-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2474030-9 1989 LTB4-omega-hydroxylase activity was inhibited (greater than 90%) by carbon monoxide or 2-diethylaminoethyl-2,2-diphenylvalerate hydrochloride (SKF-525A) (1 mM), whereas alpha-naphthoflavone produced only moderate (13%) or no effects. Carbon Monoxide 68-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-22 2474030-9 1989 LTB4-omega-hydroxylase activity was inhibited (greater than 90%) by carbon monoxide or 2-diethylaminoethyl-2,2-diphenylvalerate hydrochloride (SKF-525A) (1 mM), whereas alpha-naphthoflavone produced only moderate (13%) or no effects. Proadifen 87-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-22 2474030-9 1989 LTB4-omega-hydroxylase activity was inhibited (greater than 90%) by carbon monoxide or 2-diethylaminoethyl-2,2-diphenylvalerate hydrochloride (SKF-525A) (1 mM), whereas alpha-naphthoflavone produced only moderate (13%) or no effects. Proadifen 143-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-22 2677363-4 1989 The basic mechanism of action of terconazole, inhibition of fungal cytochrome P-450, is similar to that of the imidazoles. terconazole 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 2474030-9 1989 LTB4-omega-hydroxylase activity was inhibited (greater than 90%) by carbon monoxide or 2-diethylaminoethyl-2,2-diphenylvalerate hydrochloride (SKF-525A) (1 mM), whereas alpha-naphthoflavone produced only moderate (13%) or no effects. alpha-naphthoflavone 169-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-22 2677363-5 1989 However, because of the additional nitrogen atom in the triazole ring and the lipophilic tail, terconazole establishes a firmer and longer-lasting link with the membrane-bound fungal cytochrome P-450. Nitrogen 35-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 2677363-7 1989 Terconazole has far greater selectively for yeast cytochrome P-450 than for mammalian microsomal cytochrome P-450. terconazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 2677363-10 1989 The high affinity and selectivity of terconazole for fungal cytochrome P-450 and its potent in vitro and in vivo activity indicate that the drug may well become a significant agent for the treatment of fungal disease. terconazole 37-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 2677363-5 1989 However, because of the additional nitrogen atom in the triazole ring and the lipophilic tail, terconazole establishes a firmer and longer-lasting link with the membrane-bound fungal cytochrome P-450. Triazoles 56-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 2677363-5 1989 However, because of the additional nitrogen atom in the triazole ring and the lipophilic tail, terconazole establishes a firmer and longer-lasting link with the membrane-bound fungal cytochrome P-450. terconazole 95-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 2677363-7 1989 Terconazole has far greater selectively for yeast cytochrome P-450 than for mammalian microsomal cytochrome P-450. terconazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 2669966-1 1989 The human liver cytochrome P-450 (P-450) proteins responsible for catalyzing the oxidation of mephenytoin, tolbutamide, and hexobarbital are encoded by a multigene family (CYP2C). Mephenytoin 94-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 2804165-0 1989 [Inactivation of cytochrome P-450 by hydrogen peroxide formed in the catalytic cycle during peroxy-complex degradation]. Hydrogen Peroxide 37-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 2804165-1 1989 It was shown that the crucial role in the inactivation of microsomal cytochrome P-450 in reactions of hydroxylation of type I (DMA, AP, BPh, p-NA) and type II (AN) substrates belongs to H2O2 directly formed in the enzyme active center during the decomposition of the peroxy complex. N-myristoyl-alaninol 127-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 2804165-1 1989 It was shown that the crucial role in the inactivation of microsomal cytochrome P-450 in reactions of hydroxylation of type I (DMA, AP, BPh, p-NA) and type II (AN) substrates belongs to H2O2 directly formed in the enzyme active center during the decomposition of the peroxy complex. Hydrogen Peroxide 186-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 2512389-1 1989 The involvement of cytochrome P-450 isozymes in the activation of benzo[a]pyrene (BP) by human placental and liver microsomes was studied in vitro using monoclonal antibodies (Mab) toward the major 3-methylcholanthrene (MC)-inducible and phenobarbital-inductible rat liver P-450 isozymes (Mab 1-7-1 and Mab 2-66-3, respectively). Benzo(a)pyrene 66-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 2512389-1 1989 The involvement of cytochrome P-450 isozymes in the activation of benzo[a]pyrene (BP) by human placental and liver microsomes was studied in vitro using monoclonal antibodies (Mab) toward the major 3-methylcholanthrene (MC)-inducible and phenobarbital-inductible rat liver P-450 isozymes (Mab 1-7-1 and Mab 2-66-3, respectively). Benzo(a)pyrene 82-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 2512389-1 1989 The involvement of cytochrome P-450 isozymes in the activation of benzo[a]pyrene (BP) by human placental and liver microsomes was studied in vitro using monoclonal antibodies (Mab) toward the major 3-methylcholanthrene (MC)-inducible and phenobarbital-inductible rat liver P-450 isozymes (Mab 1-7-1 and Mab 2-66-3, respectively). Methylcholanthrene 198-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 2761265-0 1989 Specificity of steroid binding to testicular microsomal cytochrome P-450. Steroids 15-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 2761265-2 1989 On the basis of the concept that steroids accumulate in the lipid phase of endoplasmic reticulum membranes and approach the active sites of steroidogenic cytochromes P-450 from a hydrophobic environment, we describe a procedure that allows calculation of spectral dissociation constants Ks for steroid interaction with testicular microsomal cytochrome P-450 after correction for hydrophobic association of ligand with the membrane. Steroids 33-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 341-357 2761265-2 1989 On the basis of the concept that steroids accumulate in the lipid phase of endoplasmic reticulum membranes and approach the active sites of steroidogenic cytochromes P-450 from a hydrophobic environment, we describe a procedure that allows calculation of spectral dissociation constants Ks for steroid interaction with testicular microsomal cytochrome P-450 after correction for hydrophobic association of ligand with the membrane. Steroids 33-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 341-357 2508765-0 1989 [Study of the role of lysine residues of the cholesterol hydroxylating cytochrome P-450 by a method of chemical modification]. Lysine 22-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 2508765-0 1989 [Study of the role of lysine residues of the cholesterol hydroxylating cytochrome P-450 by a method of chemical modification]. Cholesterol 45-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 2765333-1 1989 Following anaesthesia with enflurane, some patients receiving isoniazid have increased serum concentrations of fluoride ion, presumably because of induction of an isozyme of cytochrome P450 which is responsible for enflurane biodegradation. Enflurane 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-189 2765333-1 1989 Following anaesthesia with enflurane, some patients receiving isoniazid have increased serum concentrations of fluoride ion, presumably because of induction of an isozyme of cytochrome P450 which is responsible for enflurane biodegradation. Isoniazid 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-189 2765333-1 1989 Following anaesthesia with enflurane, some patients receiving isoniazid have increased serum concentrations of fluoride ion, presumably because of induction of an isozyme of cytochrome P450 which is responsible for enflurane biodegradation. Fluorides 111-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-189 2765333-1 1989 Following anaesthesia with enflurane, some patients receiving isoniazid have increased serum concentrations of fluoride ion, presumably because of induction of an isozyme of cytochrome P450 which is responsible for enflurane biodegradation. Enflurane 215-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-189 2753040-0 1989 Rapid partial degradation of DDT by a cytochrome P-450 model system. DDT 29-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 2753040-1 1989 Heme compounds, in combination with a reducing agent and oxygen, can express various activities of cytochrome P-450 enzymes. Heme 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 2753040-1 1989 Heme compounds, in combination with a reducing agent and oxygen, can express various activities of cytochrome P-450 enzymes. Oxygen 57-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 2672473-0 1989 The control of cytochrome P-450 gene expression by dioxin. Dioxins 51-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 2500151-1 1989 Cytochrome P-450, designated as P-450-MK2, was purified to an electrophoretic homogeneity from polychlorinated biphenyl (PCB)-treated female crab-eating monkeys. Polychlorinated Biphenyls 95-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2500151-1 1989 Cytochrome P-450, designated as P-450-MK2, was purified to an electrophoretic homogeneity from polychlorinated biphenyl (PCB)-treated female crab-eating monkeys. Polychlorinated Biphenyls 121-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2669966-1 1989 The human liver cytochrome P-450 (P-450) proteins responsible for catalyzing the oxidation of mephenytoin, tolbutamide, and hexobarbital are encoded by a multigene family (CYP2C). Tolbutamide 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 2669966-1 1989 The human liver cytochrome P-450 (P-450) proteins responsible for catalyzing the oxidation of mephenytoin, tolbutamide, and hexobarbital are encoded by a multigene family (CYP2C). Hexobarbital 124-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 2790077-0 1989 [Immunochemical study of cholesterol-hydroxylating cytochrome P-450. Cholesterol 25-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 2706014-4 1989 Bis(tri-n-butyltin)oxide (TBTO) produced dose- and time-dependent decreases in the content and functional activity of intestinal cytochrome P-450, together with an elevation (3-fold) in the activity of microsomal heme oxygenase. bis(tri-n-butyltin)oxide 0-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 2519716-1 1989 In this perspective we have described a newly characterized pathway for the metabolism of the prosthetic heme of cytochrome P-450, which results in the formation of protein-bound adducts. Heme 105-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 2519716-2 1989 This reaction occurs when the cytochrome P-450 metabolizes a variety of xenobiotics as well as endogenous compounds such as hydrogen peroxide and lipid hydroperoxides. Hydrogen Peroxide 124-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 2519716-2 1989 This reaction occurs when the cytochrome P-450 metabolizes a variety of xenobiotics as well as endogenous compounds such as hydrogen peroxide and lipid hydroperoxides. Lipid Peroxides 146-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 2707734-1 1989 Isaxonine and several other drugs transformed by cytochrome P-450 into reactive metabolites apparently lead to immunoallergic hepatitis in man. isaxonine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 2707734-6 1989 At 60 min, the amount of isaxonine metabolite covalently bound per mg of protein was similar in plasma membranes (0.42 nmole metabolite.mg protein-1) and in microsomes (0.38); both values were decreased by about 70% in rats pretreated with piperonyl butoxide, an inhibitor of cytochrome P-450. isaxonine 25-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 276-292 2634818-6 1989 This could result from an additional induction of cytochrome P-450 caused by alcohol and an accelerated oxidation of the drugs. Alcohols 77-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 2495281-6 1989 Cytochrome P-450 reduction was also examined in both phospholipid-containing and soluble systems where the LM2 and reductase were not present as a preformed complex. Phospholipids 53-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2670357-8 1989 Specific forms of cytochrome P-450 have been shown to be involved in the initial step in the activation (N-hydroxylation) of aromatic amines and amides. aromatic amines 125-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 2670357-8 1989 Specific forms of cytochrome P-450 have been shown to be involved in the initial step in the activation (N-hydroxylation) of aromatic amines and amides. Amides 145-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 2670357-14 1989 Polymorphisms in both cytochrome P-450 and N-acetyltransferase expression may be critical determinants in the susceptibility of individuals to the toxicity and carcinogenicity of aromatic amines and amides. aromatic amines 179-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-62 2670357-14 1989 Polymorphisms in both cytochrome P-450 and N-acetyltransferase expression may be critical determinants in the susceptibility of individuals to the toxicity and carcinogenicity of aromatic amines and amides. Amides 199-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-62 2496984-0 1989 On the role of phospholipids in the reconstituted cytochrome P-450 system. Phospholipids 15-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 2496984-3 1989 The hypotheses proposed in the literature for the role of phospholipids in the reconstituted cytochrome P-450 system, mainly based on the comparison of systems without phospholipid and with Lau2GroPCho, were either validated or shown to be unlikely when tested by comparing reconstituted systems with different phosphatidylcholines. Phospholipids 58-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2496984-3 1989 The hypotheses proposed in the literature for the role of phospholipids in the reconstituted cytochrome P-450 system, mainly based on the comparison of systems without phospholipid and with Lau2GroPCho, were either validated or shown to be unlikely when tested by comparing reconstituted systems with different phosphatidylcholines. Phospholipids 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2496984-3 1989 The hypotheses proposed in the literature for the role of phospholipids in the reconstituted cytochrome P-450 system, mainly based on the comparison of systems without phospholipid and with Lau2GroPCho, were either validated or shown to be unlikely when tested by comparing reconstituted systems with different phosphatidylcholines. lau2gropcho 190-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2496984-3 1989 The hypotheses proposed in the literature for the role of phospholipids in the reconstituted cytochrome P-450 system, mainly based on the comparison of systems without phospholipid and with Lau2GroPCho, were either validated or shown to be unlikely when tested by comparing reconstituted systems with different phosphatidylcholines. Phosphatidylcholines 311-331 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2496984-4 1989 The higher activity in the Lau2GroPCho system as compared to the Ste2GroPCho system cannot be ascribed to (a) an increased affinity of cytochrome P-450 for the NADPH-cytochrome reductase in the Lau2GroPCho system, also not to (b) a Lau2GroPCho-dependent dissociation of protein multimers, nor to (c) a change in the spin state of the heme. Heme 334-338 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 2496984-7 1989 From these observations it is concluded that the higher activity of the Lau2GroPCho system compared with the Ste2GroPCho system or with a system without additional phosphatidylcholine may at least in part be caused by a difference in the conformation of the cytochrome P-450 molecules in these systems. Phosphatidylcholines 164-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-274 2496984-8 1989 Furthermore, the different effects of both phosphatidylcholines on the Km and V for pentoxyresorufin not only suggest a role of phospholipids in the binding of the substrate to the active site of the cytochrome P-450 molecule, but also on the efficiency of electron transfer from NADPH-cytochrome reductase to cytochrome P-450. Phosphatidylcholines 43-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-216 2496984-8 1989 Furthermore, the different effects of both phosphatidylcholines on the Km and V for pentoxyresorufin not only suggest a role of phospholipids in the binding of the substrate to the active site of the cytochrome P-450 molecule, but also on the efficiency of electron transfer from NADPH-cytochrome reductase to cytochrome P-450. Phosphatidylcholines 43-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 310-326 2496984-8 1989 Furthermore, the different effects of both phosphatidylcholines on the Km and V for pentoxyresorufin not only suggest a role of phospholipids in the binding of the substrate to the active site of the cytochrome P-450 molecule, but also on the efficiency of electron transfer from NADPH-cytochrome reductase to cytochrome P-450. pentoxyresorufin 84-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-216 2496984-8 1989 Furthermore, the different effects of both phosphatidylcholines on the Km and V for pentoxyresorufin not only suggest a role of phospholipids in the binding of the substrate to the active site of the cytochrome P-450 molecule, but also on the efficiency of electron transfer from NADPH-cytochrome reductase to cytochrome P-450. pentoxyresorufin 84-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 310-326 2496984-8 1989 Furthermore, the different effects of both phosphatidylcholines on the Km and V for pentoxyresorufin not only suggest a role of phospholipids in the binding of the substrate to the active site of the cytochrome P-450 molecule, but also on the efficiency of electron transfer from NADPH-cytochrome reductase to cytochrome P-450. Phospholipids 128-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-216 2496984-8 1989 Furthermore, the different effects of both phosphatidylcholines on the Km and V for pentoxyresorufin not only suggest a role of phospholipids in the binding of the substrate to the active site of the cytochrome P-450 molecule, but also on the efficiency of electron transfer from NADPH-cytochrome reductase to cytochrome P-450. Phospholipids 128-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 310-326 2758074-6 1989 The incorporation of cytochrome P-450 into liposomes caused the acceleration of PE and PG hydrolysis. Phosphatidylglycerols 87-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 2702801-6 1989 This would imply that the cytochrome P-450 determined by the debrisoquin allele or some coinherited 4-hydroxylase(s) was induced to a greater extent in EMs than PMs. Debrisoquin 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2706014-4 1989 Bis(tri-n-butyltin)oxide (TBTO) produced dose- and time-dependent decreases in the content and functional activity of intestinal cytochrome P-450, together with an elevation (3-fold) in the activity of microsomal heme oxygenase. bis(tri-n-butyltin)oxide 26-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 2706014-5 1989 The effects of di-n-butyltin dichloride on heme oxygenase and cytochrome P-450 were pronounced in the small intestine and extended to the liver and kidneys within 21 hr after oral-exposure, whereas TBTO did not affect the liver until much later (6 days), when cytochrome P-450 content was reduced markedly (30%). dibutyldichlorotin 15-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 2706014-5 1989 The effects of di-n-butyltin dichloride on heme oxygenase and cytochrome P-450 were pronounced in the small intestine and extended to the liver and kidneys within 21 hr after oral-exposure, whereas TBTO did not affect the liver until much later (6 days), when cytochrome P-450 content was reduced markedly (30%). dibutyldichlorotin 15-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 260-276 2651504-4 1989 The dialkylnitrosamine carcinogens are activated by NADPH-dependent cytochrome P-450 enzymes in their target tissues. dialkylnitrosamine 4-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2512166-4 1989 These findings suggest a certain inability of cytochrome P-450 mono-oxygenases to metabolise a specific group of PCBs. Polychlorinated Biphenyls 113-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2649791-1 1989 A cDNA of a human liver cytochrome P-450, corresponding to P-450 human-2, was expressed in Saccharomyces cerevisiae cells by the use of a galactose-inducible expression vector containing the GAL7 promoter and terminator. Galactose 138-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-72 2693891-0 1989 Induction of hepatic cytochrome P450 gene expression by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Polychlorinated Dibenzodioxins 56-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 2739204-5 1989 Isoprothiolane did not promote the activity of the demethylase and hydroxylase in liver microsomes but it promoted the formation of cytochrome P-450 and b5 whose contents in the animals treated with isoprothiolane of a dose of 50 or 100 mg/kg were about twice as high as those in the non-treated animals. isoprothiolane 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 2739204-5 1989 Isoprothiolane did not promote the activity of the demethylase and hydroxylase in liver microsomes but it promoted the formation of cytochrome P-450 and b5 whose contents in the animals treated with isoprothiolane of a dose of 50 or 100 mg/kg were about twice as high as those in the non-treated animals. isoprothiolane 199-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 2930594-2 1989 Under conditions enabling cytochrome P-450-dependent benzylic hydroxylation of BPBS and BDVS, both benzyl sulfides were mutagenic. bpbs 79-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2930594-2 1989 Under conditions enabling cytochrome P-450-dependent benzylic hydroxylation of BPBS and BDVS, both benzyl sulfides were mutagenic. bdvs 88-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2930594-2 1989 Under conditions enabling cytochrome P-450-dependent benzylic hydroxylation of BPBS and BDVS, both benzyl sulfides were mutagenic. benzyl sulfide 99-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2930595-0 1989 Inhibitory effects of nilutamide, a new androgen receptor antagonist, on mouse and human liver cytochrome P-450. nilutamide 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 2930595-12 1989 These results show that nilutamide inhibits hepatic cytochrome P-450 activity, and suggest that inhibition may actually occur after therapeutic doses of nilutamide in humans. nilutamide 24-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 2651504-4 1989 The dialkylnitrosamine carcinogens are activated by NADPH-dependent cytochrome P-450 enzymes in their target tissues. NADP 52-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2917971-2 1989 Cytochrome P-450 is known to catalyze the following oxygen transfer reaction: RH + PhIO----ROH + PhI where RH represents a variety of hydroxylatable substrates and PhIO a variety of iodosobenzene derivatives that serve as oxygen donors, and neither molecular oxygen nor an external electron donor is required. Oxygen 52-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2651504-7 1989 We have demonstrated that dietary zinc deficiency increased the cytochrome P-450-dependent microsomal metabolism of methylbenzylnitrosamine and dimethylnitrosamine, two members of this class of dialkylnitrosamine carcinogens, while the addition of zinc in vitro noncompetitively inhibits the microsomal metabolism of these carcinogens. nitrosobenzylmethylamine 116-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 2917971-2 1989 Cytochrome P-450 is known to catalyze the following oxygen transfer reaction: RH + PhIO----ROH + PhI where RH represents a variety of hydroxylatable substrates and PhIO a variety of iodosobenzene derivatives that serve as oxygen donors, and neither molecular oxygen nor an external electron donor is required. iodosobenzene 182-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2651504-7 1989 We have demonstrated that dietary zinc deficiency increased the cytochrome P-450-dependent microsomal metabolism of methylbenzylnitrosamine and dimethylnitrosamine, two members of this class of dialkylnitrosamine carcinogens, while the addition of zinc in vitro noncompetitively inhibits the microsomal metabolism of these carcinogens. Dimethylnitrosamine 144-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 2917971-2 1989 Cytochrome P-450 is known to catalyze the following oxygen transfer reaction: RH + PhIO----ROH + PhI where RH represents a variety of hydroxylatable substrates and PhIO a variety of iodosobenzene derivatives that serve as oxygen donors, and neither molecular oxygen nor an external electron donor is required. Oxygen 222-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2917971-2 1989 Cytochrome P-450 is known to catalyze the following oxygen transfer reaction: RH + PhIO----ROH + PhI where RH represents a variety of hydroxylatable substrates and PhIO a variety of iodosobenzene derivatives that serve as oxygen donors, and neither molecular oxygen nor an external electron donor is required. Oxygen 222-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2651504-7 1989 We have demonstrated that dietary zinc deficiency increased the cytochrome P-450-dependent microsomal metabolism of methylbenzylnitrosamine and dimethylnitrosamine, two members of this class of dialkylnitrosamine carcinogens, while the addition of zinc in vitro noncompetitively inhibits the microsomal metabolism of these carcinogens. dialkylnitrosamine 194-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 2565211-2 1989 Purification and identification of the rifampicin-inducible human liver cytochrome P-450 (cyclosporin A oxidase) as a product of P450IIIA gene subfamily. Rifampin 39-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 2752065-0 1989 [Interaction of cholesterol hydroxylating cytochrome P-450 with cytochrome b5]. Cholesterol 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 2565211-3 1989 A cytochrome P-450 involved in the metabolism of cyclosporin A (CsA) was isolated and purified to electrophoretic homogeneity from human liver microsomes of renal transplant donors. Cyclosporine 49-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-18 2565211-3 1989 A cytochrome P-450 involved in the metabolism of cyclosporin A (CsA) was isolated and purified to electrophoretic homogeneity from human liver microsomes of renal transplant donors. Cyclosporine 64-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-18 2650039-4 1989 Chloroform is discussed as an example of cytochrome P-450-mediated activation and dihaloethanes and hexachloro-1,3-butadiene as examples of glutathione conjugation followed by activation. Chloroform 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 2744145-0 1989 [The immunotropic activity of GABA during cytochrome P-450 induction in the liver]. gamma-Aminobutyric Acid 30-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 2744145-2 1989 The use of GABA under induction of cytochrome P-450 of the liver by phenobarbital or rifampicin was not followed by immunodepression. gamma-Aminobutyric Acid 11-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 2744145-2 1989 The use of GABA under induction of cytochrome P-450 of the liver by phenobarbital or rifampicin was not followed by immunodepression. Phenobarbital 68-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 2744145-2 1989 The use of GABA under induction of cytochrome P-450 of the liver by phenobarbital or rifampicin was not followed by immunodepression. Rifampin 85-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 2744145-3 1989 The obtained data indicate the dependence of GABA pharmacological activity from the function of cytochrome P-450-dependent monooxygenase system of the liver. gamma-Aminobutyric Acid 45-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 2703963-1 1989 Acetaminophen (AA) is converted, presumably by cytochrome P-450, to an electrophile which is conjugated with glutathione (GS). Acetaminophen 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 2703963-6 1989 Of the cytochrome P-450 inducers, 3-methylcholanthrene and 2,3,7,8-tetrachlorodibenzo-p-dioxin, increased the biliary excretion of AA-GS (2.9- and 3.2-fold, respectively) whereas ethanol and isoniazid did not affect it, and pregnenolone-16 alpha-carbonitrile tended to decrease it (43%). Methylcholanthrene 34-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 2703963-6 1989 Of the cytochrome P-450 inducers, 3-methylcholanthrene and 2,3,7,8-tetrachlorodibenzo-p-dioxin, increased the biliary excretion of AA-GS (2.9- and 3.2-fold, respectively) whereas ethanol and isoniazid did not affect it, and pregnenolone-16 alpha-carbonitrile tended to decrease it (43%). Polychlorinated Dibenzodioxins 59-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 2703963-6 1989 Of the cytochrome P-450 inducers, 3-methylcholanthrene and 2,3,7,8-tetrachlorodibenzo-p-dioxin, increased the biliary excretion of AA-GS (2.9- and 3.2-fold, respectively) whereas ethanol and isoniazid did not affect it, and pregnenolone-16 alpha-carbonitrile tended to decrease it (43%). Ethanol 179-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 2703963-6 1989 Of the cytochrome P-450 inducers, 3-methylcholanthrene and 2,3,7,8-tetrachlorodibenzo-p-dioxin, increased the biliary excretion of AA-GS (2.9- and 3.2-fold, respectively) whereas ethanol and isoniazid did not affect it, and pregnenolone-16 alpha-carbonitrile tended to decrease it (43%). Isoniazid 191-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 2703963-6 1989 Of the cytochrome P-450 inducers, 3-methylcholanthrene and 2,3,7,8-tetrachlorodibenzo-p-dioxin, increased the biliary excretion of AA-GS (2.9- and 3.2-fold, respectively) whereas ethanol and isoniazid did not affect it, and pregnenolone-16 alpha-carbonitrile tended to decrease it (43%). Pregnenolone 224-236 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 2703963-6 1989 Of the cytochrome P-450 inducers, 3-methylcholanthrene and 2,3,7,8-tetrachlorodibenzo-p-dioxin, increased the biliary excretion of AA-GS (2.9- and 3.2-fold, respectively) whereas ethanol and isoniazid did not affect it, and pregnenolone-16 alpha-carbonitrile tended to decrease it (43%). 16 alpha-carbonitrile 237-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 2703963-8 1989 Several cytochrome P-450 inhibitors [metyrapone, 8-methoxypsoralen, 2-(4,6-dichloro-biphenyloxy) ethylamine, alpha-naphthoflavone and cimetidine] decreased the biliary excretion of AA-GS, although SKF 525-A and piperonyl butoxide did not. Methoxsalen 49-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2703963-8 1989 Several cytochrome P-450 inhibitors [metyrapone, 8-methoxypsoralen, 2-(4,6-dichloro-biphenyloxy) ethylamine, alpha-naphthoflavone and cimetidine] decreased the biliary excretion of AA-GS, although SKF 525-A and piperonyl butoxide did not. 2-(4,6-dichloro-biphenyloxy) ethylamine 68-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2703963-8 1989 Several cytochrome P-450 inhibitors [metyrapone, 8-methoxypsoralen, 2-(4,6-dichloro-biphenyloxy) ethylamine, alpha-naphthoflavone and cimetidine] decreased the biliary excretion of AA-GS, although SKF 525-A and piperonyl butoxide did not. alpha-naphthoflavone 109-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2703963-8 1989 Several cytochrome P-450 inhibitors [metyrapone, 8-methoxypsoralen, 2-(4,6-dichloro-biphenyloxy) ethylamine, alpha-naphthoflavone and cimetidine] decreased the biliary excretion of AA-GS, although SKF 525-A and piperonyl butoxide did not. Proadifen 197-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2703963-8 1989 Several cytochrome P-450 inhibitors [metyrapone, 8-methoxypsoralen, 2-(4,6-dichloro-biphenyloxy) ethylamine, alpha-naphthoflavone and cimetidine] decreased the biliary excretion of AA-GS, although SKF 525-A and piperonyl butoxide did not. Piperonyl Butoxide 211-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2713464-0 1989 [Tetraphenylporphyrin-Sn4+ induction of cytochrome P-450]. tin tetraphenylporphyrin 1-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-57 2713464-5 1989 To clarity the mechanism of TPP-Sn4+ effect on cytochrome P-450, we studied its effect on the activity of heme oxygenase and LP rate. tpp-sn4 28-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 2644935-3 1989 CDDP induces a dramatic change in both the testosterone level and the microsomal cytochrome P-450 concentration. cddp 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 2914926-0 1989 19F nuclear magnetic resonance as a probe of the spatial relationship between the heme iron of cytochrome P-450 and its substrate. Heme 82-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 2646846-1 1989 Hepatic cytochrome-P-450-linked microsomal metabolism is inhibited by cimetidine, and to a lesser extent by ranitidine. Cimetidine 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2914926-0 1989 19F nuclear magnetic resonance as a probe of the spatial relationship between the heme iron of cytochrome P-450 and its substrate. Iron 87-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 2914926-1 1989 The distance between the heme iron of ferrous cytochrome P-450-CAM and a fluorine label attached to the 9-methyl carbon of its substrate, (1R)-(+)-camphor, has been determined using 19F NMR. Heme 25-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2914926-1 1989 The distance between the heme iron of ferrous cytochrome P-450-CAM and a fluorine label attached to the 9-methyl carbon of its substrate, (1R)-(+)-camphor, has been determined using 19F NMR. Iron 30-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2917006-6 1989 Both the increase and the decrease in cytochrome P-450 could be prevented by the administration of the protein synthesis inhibitor puromycin. Puromycin 131-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 2917006-7 1989 Various isozymes of cytochrome P-450 induced by phenobarbital, beta-napthaflavone and clofibrate were also depressed by this interferon. Phenobarbital 48-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 2917006-7 1989 Various isozymes of cytochrome P-450 induced by phenobarbital, beta-napthaflavone and clofibrate were also depressed by this interferon. beta-napthaflavone 63-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 2917006-7 1989 Various isozymes of cytochrome P-450 induced by phenobarbital, beta-napthaflavone and clofibrate were also depressed by this interferon. Clofibrate 86-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 2923975-0 1989 [Protective action of fat- and water-soluble antioxidants on the cytochrome P-450 system during lipid peroxidation in liver microsomes]. Water 31-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 2923975-2 1989 It was shown that for optimal protection effect of cytochrome P-450 system the mixture of water and liposoluble antioxidants is required. Water 90-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 2912586-0 1989 Activation of the food-derived mutagen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine by rabbit and human liver microsomes and purified forms of cytochrome P-450. 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine 39-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-162 2914926-1 1989 The distance between the heme iron of ferrous cytochrome P-450-CAM and a fluorine label attached to the 9-methyl carbon of its substrate, (1R)-(+)-camphor, has been determined using 19F NMR. Fluorine 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2914926-1 1989 The distance between the heme iron of ferrous cytochrome P-450-CAM and a fluorine label attached to the 9-methyl carbon of its substrate, (1R)-(+)-camphor, has been determined using 19F NMR. Carbon 113-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2914926-1 1989 The distance between the heme iron of ferrous cytochrome P-450-CAM and a fluorine label attached to the 9-methyl carbon of its substrate, (1R)-(+)-camphor, has been determined using 19F NMR. Camphor 138-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2663083-4 1989 Besides, some enzymes catalyzing the interconversions of active oxygen species (catalase superoxide dismutase, cytochrome P-450) are also inactivated in the course of catalysis under the oxidative action of active oxygen species. Oxygen 64-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 2663083-4 1989 Besides, some enzymes catalyzing the interconversions of active oxygen species (catalase superoxide dismutase, cytochrome P-450) are also inactivated in the course of catalysis under the oxidative action of active oxygen species. Oxygen 214-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 2646846-1 1989 Hepatic cytochrome-P-450-linked microsomal metabolism is inhibited by cimetidine, and to a lesser extent by ranitidine. Ranitidine 108-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2546388-0 1989 Corneal arachidonate metabolism via cytochrome P450: characterization of two novel biologically active metabolites. Arachidonic Acid 8-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 2784911-0 1989 Role of kidney microsomal cytochrome P-450 in cyclosporin induced nephropathy. Cyclosporine 46-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2785364-0 1989 Studies on the interactions of MPTP(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) with the cytochrome P-450 enzyme system--clues to a possible aetiological factor in Parkinson"s disease. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 31-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 2565119-1 1989 The H2-receptor antagonist cimetidine has been reported to decrease the hepatic clearance of numerous drugs by inhibiting cytochrome P-450 metabolism, decreasing liver blood flow or both. Cimetidine 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 2519233-0 1989 A theoretical study on the metabolic activation of paracetamol by cytochrome P-450: indications for a uniform oxidation mechanism. Acetaminophen 51-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 2519233-1 1989 The cytochrome P-450 mediated activation of paracetamol (PAR) to the reactive electrophilic intermediate N-acetyl-p-benzoquinone imine (NAPQI) has been studied by use of SV 6-31G ab initio energy calculations and spin distributions. Acetaminophen 44-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2519233-1 1989 The cytochrome P-450 mediated activation of paracetamol (PAR) to the reactive electrophilic intermediate N-acetyl-p-benzoquinone imine (NAPQI) has been studied by use of SV 6-31G ab initio energy calculations and spin distributions. Acetaminophen 57-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2519233-1 1989 The cytochrome P-450 mediated activation of paracetamol (PAR) to the reactive electrophilic intermediate N-acetyl-p-benzoquinone imine (NAPQI) has been studied by use of SV 6-31G ab initio energy calculations and spin distributions. N-acetyl-4-benzoquinoneimine 105-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2519233-1 1989 The cytochrome P-450 mediated activation of paracetamol (PAR) to the reactive electrophilic intermediate N-acetyl-p-benzoquinone imine (NAPQI) has been studied by use of SV 6-31G ab initio energy calculations and spin distributions. N-acetyl-4-benzoquinoneimine 136-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2519233-2 1989 A simplified model for cytochrome P-450 has been used by substituting the proposed biologically active ferric-oxene state of cytochrome P-450 by a singlet oxygen atom. ferric-oxene 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 2519233-2 1989 A simplified model for cytochrome P-450 has been used by substituting the proposed biologically active ferric-oxene state of cytochrome P-450 by a singlet oxygen atom. ferric-oxene 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 2519233-2 1989 A simplified model for cytochrome P-450 has been used by substituting the proposed biologically active ferric-oxene state of cytochrome P-450 by a singlet oxygen atom. Oxygen 155-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 2515836-6 1989 However, noxious effects may instead be induced for chemicals bioactivated by cytochrome P-450-dependent reactions, such as N-nitrosamines. n-nitrosamines 124-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 2496778-0 1989 Cardiovascular and renal actions of cytochrome P-450 metabolites of arachidonic acid. Arachidonic Acid 68-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 2496778-2 1989 Arachidonic acid (AA) can be oxidatively metabolized by cytochrome P-450 hemoproteins to an array of compounds identified as constituents of tissues and biological fluids. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 2785364-0 1989 Studies on the interactions of MPTP(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) with the cytochrome P-450 enzyme system--clues to a possible aetiological factor in Parkinson"s disease. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 36-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 2785364-2 1989 This study examines the interactions of MPTP with the cytochrome P-450 system--an enzyme system which is known to be involved in the detoxication of MPTP. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 40-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 2785364-2 1989 This study examines the interactions of MPTP with the cytochrome P-450 system--an enzyme system which is known to be involved in the detoxication of MPTP. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 149-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 2598298-2 1989 Both a reconstituted system of cytochrome P-450 (P-450) and cytochrome b5 (b5) with NADPH was capable of reducing Na2CrO4 (30 microM) provided anaerobic atmosphere. NADP 84-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 2598298-2 1989 Both a reconstituted system of cytochrome P-450 (P-450) and cytochrome b5 (b5) with NADPH was capable of reducing Na2CrO4 (30 microM) provided anaerobic atmosphere. na2cro4 114-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 2785364-6 1989 MPTP was found from in vitro studies to cause a mixed non-competitive inhibition of cytochrome P-450 dependent ethoxyresorufin O-dealkylase activity with an inhibition constant (Ki) of 0.06 mM. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 2642041-0 1989 Cytochrome P-450-catalyzed asymmetric epoxidation of simple prochiral and chiral aliphatic alkenes: species dependence and effect of enzyme induction on enantioselective oxirane formation. Alkenes 91-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2785364-7 1989 Two binding sites of MPTP to hepatic cytochrome P-450 were found by spectral perturbation studies--the higher affinity site binding about a hundred times more avidly to MPTP than the other. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 21-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 2642041-0 1989 Cytochrome P-450-catalyzed asymmetric epoxidation of simple prochiral and chiral aliphatic alkenes: species dependence and effect of enzyme induction on enantioselective oxirane formation. Ethylene Oxide 170-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2785364-7 1989 Two binding sites of MPTP to hepatic cytochrome P-450 were found by spectral perturbation studies--the higher affinity site binding about a hundred times more avidly to MPTP than the other. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 169-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 2785364-9 1989 Cytochrome P-450 levels were lowest 3 to 6 hours after treatment with MPTP. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 70-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2785364-10 1989 MPTP was also found to cause a dose-dependent decrease in the cytochrome P-450 enzyme activities bufuralol hydroxylase (buf) and aryl hydrocarbon hydroxylase (AHH) in the brain. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 2910635-0 1989 In vitro characterization of the human cytochrome P-450 involved in polymorphic oxidation of propafenone. Propafenone 93-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 2910635-10 1989 Thus it can be concluded that 5-hydroxypropafenone is formed by the cytochrome P-450 isozyme involved in polymorphic bufuralol oxidation. 5-hydroxypropafenone 30-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2910635-10 1989 Thus it can be concluded that 5-hydroxypropafenone is formed by the cytochrome P-450 isozyme involved in polymorphic bufuralol oxidation. bufuralol 117-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2910639-1 1989 The relative contributions of the debrisoquin and mephenytoin isozymes of hepatic cytochrome P-450 to the stereoselective metabolism of propranolol have been studied in a panel of volunteers of known oxidative phenotypes. Debrisoquin 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 2910639-1 1989 The relative contributions of the debrisoquin and mephenytoin isozymes of hepatic cytochrome P-450 to the stereoselective metabolism of propranolol have been studied in a panel of volunteers of known oxidative phenotypes. Mephenytoin 50-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 2910639-1 1989 The relative contributions of the debrisoquin and mephenytoin isozymes of hepatic cytochrome P-450 to the stereoselective metabolism of propranolol have been studied in a panel of volunteers of known oxidative phenotypes. Propranolol 136-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 2680407-0 1989 Induction of the alcohol-inducible form of cytochrome P-450 by nitrogen-containing heterocycles: effects on pyridine N-oxide production. Nitrogen 63-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2680407-0 1989 Induction of the alcohol-inducible form of cytochrome P-450 by nitrogen-containing heterocycles: effects on pyridine N-oxide production. pyridine N-oxide 108-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2680378-0 1989 Radical intermediates in the cytochrome P-450-catalyzed oxidation of aliphatic hydrocarbons. radical 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 2680378-0 1989 Radical intermediates in the cytochrome P-450-catalyzed oxidation of aliphatic hydrocarbons. Hydrocarbons 79-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 2680385-1 1989 A variety of organic compounds such as carbon tetrachloride, halothane, gentian violet, and benznidazole are reductively metabolized to free radicals by liver microsomal cytochrome P-450. Carbon Tetrachloride 39-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-186 2698318-2 1989 Evidence is presented for the direct involvement of certain cytochrome P-450 isoenzymes in the major toxic effect of hexachlorobenzene, hepatic porphyria. Hexachlorobenzene 117-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 2680385-1 1989 A variety of organic compounds such as carbon tetrachloride, halothane, gentian violet, and benznidazole are reductively metabolized to free radicals by liver microsomal cytochrome P-450. Halothane 61-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-186 2680385-1 1989 A variety of organic compounds such as carbon tetrachloride, halothane, gentian violet, and benznidazole are reductively metabolized to free radicals by liver microsomal cytochrome P-450. Gentian Violet 72-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-186 2908851-0 1989 Metabolic oxidation of the carcinogens 4-aminobiphenyl and 4,4"-methylene-bis(2-chloroaniline) by human hepatic microsomes and by purified rat hepatic cytochrome P-450 monooxygenases. 4-biphenylamine 39-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 2908851-0 1989 Metabolic oxidation of the carcinogens 4-aminobiphenyl and 4,4"-methylene-bis(2-chloroaniline) by human hepatic microsomes and by purified rat hepatic cytochrome P-450 monooxygenases. Methylenebis(chloroaniline) 59-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 2680385-1 1989 A variety of organic compounds such as carbon tetrachloride, halothane, gentian violet, and benznidazole are reductively metabolized to free radicals by liver microsomal cytochrome P-450. benzonidazole 92-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-186 2806064-0 1989 [Rotational diffusion of cytochrome P-450 in a model membrane of various phospholipid composition]. Phospholipids 73-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 2680407-0 1989 Induction of the alcohol-inducible form of cytochrome P-450 by nitrogen-containing heterocycles: effects on pyridine N-oxide production. Alcohols 17-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2908851-7 1989 MOCA ring-oxidation and methylene carbon oxidation showed varied cytochrome P-450 selectivity and accounted for 14 to 79% of total oxidation products. Carbon 34-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 2908851-11 1989 Moreover, both the ABP N-oxidation and phenacetin O-deethylation activities of human liver microsomes showed a good correlation (R = 0.72) with the levels of cytochrome P-450 immunochemically related to rat P-450ISF-G. Phenacetin 39-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-174 2500346-3 1989 Cytochrome P-450 activity was monitored by measuring urinary 6-beta-hydroxycortisol output and systemic antipyrine clearance. 6 beta-hydroxycortisol 61-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2500346-6 1989 The results are in keeping with the well known heterogeneity of the hepatic monooxygenase system, as phenytoin and carbamazepine induce different panels of cytochrome P-450 isoenzymes. Phenytoin 101-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 2500346-6 1989 The results are in keeping with the well known heterogeneity of the hepatic monooxygenase system, as phenytoin and carbamazepine induce different panels of cytochrome P-450 isoenzymes. Carbamazepine 115-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 2570698-1 1989 The sparteine/debrisoquine oxidation polymorphism results from differences in the activity of one isozyme of cytochrome P450, the P450db1 (P450 IID1). Sparteine 4-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 2570698-1 1989 The sparteine/debrisoquine oxidation polymorphism results from differences in the activity of one isozyme of cytochrome P450, the P450db1 (P450 IID1). Debrisoquin 14-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 2634093-4 1989 This prediction is achieved by comparing the molecular conformation of these drugs with lanosterol, the substrate of the fungal cytochrome P-450, and with phenobarbitone, an inducing agent of a family of mammalian cytochrome P-450, toward which the antifungal agents show highest inhibitory activity. Phenobarbital 155-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-230 2643542-1 1989 Because steroids reach high concentrations within the adrenal cortex, effects of the direct interaction of steroids and cytochrome P450 enzymes are possible and may involve oxidative damage. Steroids 8-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 2643542-3 1989 Release of oxidants during pseudosubstrate interaction with cytochrome P450s may be responsible for loss of enzymatic activity observed; enzyme activity can be protected by cytochrome P450 inhibitors, antioxidants, and lowered oxygen concentration. Oxygen 227-233 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 2643542-5 1989 Cytochrome P450/pseudosubstrate effects could be involved in the aging and death of adrenocortical cells in vivo, and necrosis of the adrenal cortex due to excessive ACTH stimulation or due to the action of adrenolytic chemicals could result from damage by oxygen radicals originating from cytochrome P450s. Reactive Oxygen Species 257-272 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 2634093-0 1989 Adverse reactions of imidazole antifungal agents: computer graphic studies of cytochrome P-450 interactions. imidazole 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 2634093-3 1989 A computer graphic technique capable of predicting the interaction of these structurally-related imidazoles with fungal and mammalian cytochrome P-450 proteins is described. Imidazoles 97-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 2634093-4 1989 This prediction is achieved by comparing the molecular conformation of these drugs with lanosterol, the substrate of the fungal cytochrome P-450, and with phenobarbitone, an inducing agent of a family of mammalian cytochrome P-450, toward which the antifungal agents show highest inhibitory activity. Lanosterol 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 2634093-4 1989 This prediction is achieved by comparing the molecular conformation of these drugs with lanosterol, the substrate of the fungal cytochrome P-450, and with phenobarbitone, an inducing agent of a family of mammalian cytochrome P-450, toward which the antifungal agents show highest inhibitory activity. Lanosterol 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-230 2615529-10 1989 The cytochrome P-450-dependent hydroxylation capacity was evaluated with debrisoquine but no correlation was found to the pharmacokinetics of 6-MP. Debrisoquin 73-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2848247-3 1988 This gene is clearly a member of the cytochrome P-450 gene superfamily, because the sequence contains regions of marked homology to those of other members, notably a putative membrane-spanning region, I helix, Ozols, and heme-binding regions. Heme 221-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 2763709-1 1989 Oral therapy with nizoral and griseofulvin influences the system of cytochrome P = 450-dependent monoxygenases of the liver, this resulting in the depression of antipyrine microsomal oxidation. Ketoconazole 18-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-86 2763709-1 1989 Oral therapy with nizoral and griseofulvin influences the system of cytochrome P = 450-dependent monoxygenases of the liver, this resulting in the depression of antipyrine microsomal oxidation. Griseofulvin 30-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-86 3254979-2 1988 Even at a low dose (10 mg/kg), MCZ significantly increased cytochrome P-450 content and reduced nicotinamide adenine dinucleotide phosphate cytochrome c reductase activity. Miconazole 31-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 3254979-5 1988 These results suggest that MCZ, but not KCZ, has inducing activity for hepatic cytochrome P-450-dependent oxidative metabolism of steroids and xenobiotics, in addition to its known inhibitory activity. Miconazole 27-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 3254979-5 1988 These results suggest that MCZ, but not KCZ, has inducing activity for hepatic cytochrome P-450-dependent oxidative metabolism of steroids and xenobiotics, in addition to its known inhibitory activity. Steroids 130-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 3072574-0 1988 Cytochrome P-450 metabolism of arachidonic acid: formation and biological actions of "epoxygenase"-derived eicosanoids. Arachidonic Acid 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3072574-0 1988 Cytochrome P-450 metabolism of arachidonic acid: formation and biological actions of "epoxygenase"-derived eicosanoids. Eicosanoids 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3198602-2 1988 Cytochrome P-450cam normally catalyzes the regiospecific hydroxylation of the monoterpene camphor. Monoterpenes 78-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3198602-2 1988 Cytochrome P-450cam normally catalyzes the regiospecific hydroxylation of the monoterpene camphor. Camphor 90-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3198602-10 1988 Camphane bound to cytochrome P-450cam exhibits a larger decrease in high spin fraction (45%) and a correspondingly larger KD (46 microM), suggesting that the carbonyl moiety of camphor plays an important steric role in addition to its interaction as a hydrogen bond acceptor. camphane 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 3198602-10 1988 Camphane bound to cytochrome P-450cam exhibits a larger decrease in high spin fraction (45%) and a correspondingly larger KD (46 microM), suggesting that the carbonyl moiety of camphor plays an important steric role in addition to its interaction as a hydrogen bond acceptor. Camphor 177-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 3198602-10 1988 Camphane bound to cytochrome P-450cam exhibits a larger decrease in high spin fraction (45%) and a correspondingly larger KD (46 microM), suggesting that the carbonyl moiety of camphor plays an important steric role in addition to its interaction as a hydrogen bond acceptor. Hydrogen 252-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 2848247-5 1988 The expressed protein was similar in size to human placental aromatase system cytochrome P-450, as detected by immunoblot analysis, and catalyzed the aromatization of androstenedione, testosterone, and 16 alpha-hydroxyandrostenedione. Androstenedione 167-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 2848247-5 1988 The expressed protein was similar in size to human placental aromatase system cytochrome P-450, as detected by immunoblot analysis, and catalyzed the aromatization of androstenedione, testosterone, and 16 alpha-hydroxyandrostenedione. Testosterone 184-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 2848247-5 1988 The expressed protein was similar in size to human placental aromatase system cytochrome P-450, as detected by immunoblot analysis, and catalyzed the aromatization of androstenedione, testosterone, and 16 alpha-hydroxyandrostenedione. alpha-hydroxyandrostenedione 205-233 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 3186722-4 1988 Using two assay systems with different selectivity for the two cytochrome P-450 isozymes catalyzing bufuralol metabolism in human liver, we show that anti-LKM1 exclusively recognizes cytochrome P-450db1. bufuralol 100-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 3196735-2 1988 19-Hydroxy-PGE1 and 19-hydroxy-PGE2 are formed from PGE1 and PGE2 by prostaglandin 19-hydroxylase, a cytochrome P-450 enzyme, in seminal vesicles. 19-hydroxyprostaglandin E1 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-117 3196735-2 1988 19-Hydroxy-PGE1 and 19-hydroxy-PGE2 are formed from PGE1 and PGE2 by prostaglandin 19-hydroxylase, a cytochrome P-450 enzyme, in seminal vesicles. 19-hydroxyprostaglandin E2 20-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-117 3196735-2 1988 19-Hydroxy-PGE1 and 19-hydroxy-PGE2 are formed from PGE1 and PGE2 by prostaglandin 19-hydroxylase, a cytochrome P-450 enzyme, in seminal vesicles. Alprostadil 11-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-117 3196735-2 1988 19-Hydroxy-PGE1 and 19-hydroxy-PGE2 are formed from PGE1 and PGE2 by prostaglandin 19-hydroxylase, a cytochrome P-450 enzyme, in seminal vesicles. Dinoprostone 31-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-117 3196735-7 1988 20-Hydroxy-PGE1 and 20-hydroxy-PGE2 are formed from PGE1 and PGE2 by cytochrome P-450 in the vesicular glands and the ampullae of deferent ducts of the ram. 20-hydroxyprostaglandin E1 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3196735-7 1988 20-Hydroxy-PGE1 and 20-hydroxy-PGE2 are formed from PGE1 and PGE2 by cytochrome P-450 in the vesicular glands and the ampullae of deferent ducts of the ram. 20-hydroxyprostaglandin E2 20-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3196735-7 1988 20-Hydroxy-PGE1 and 20-hydroxy-PGE2 are formed from PGE1 and PGE2 by cytochrome P-450 in the vesicular glands and the ampullae of deferent ducts of the ram. Alprostadil 11-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3196735-7 1988 20-Hydroxy-PGE1 and 20-hydroxy-PGE2 are formed from PGE1 and PGE2 by cytochrome P-450 in the vesicular glands and the ampullae of deferent ducts of the ram. Dinoprostone 31-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3196735-9 1988 Polymorphism of cytochrome P-450 might contribute to variations in seminal prostaglandins in man and in sheep. Prostaglandins 75-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 3196361-9 1988 For a better understanding of the cytochrome P-450 mediated reactions, we studied the metabolism of midazolam in microsomal fractions prepared from twelve human livers. Midazolam 100-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 3196361-11 1988 Furthermore, and since we demonstrated that midazolam was predominantly transformed by a single cytochrome P-450 enzyme, we could attribute the large inter-individual variations in midazolam metabolism to differences in human liver cytochrome P-450 content. Midazolam 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 3196361-11 1988 Furthermore, and since we demonstrated that midazolam was predominantly transformed by a single cytochrome P-450 enzyme, we could attribute the large inter-individual variations in midazolam metabolism to differences in human liver cytochrome P-450 content. Midazolam 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-248 3196361-11 1988 Furthermore, and since we demonstrated that midazolam was predominantly transformed by a single cytochrome P-450 enzyme, we could attribute the large inter-individual variations in midazolam metabolism to differences in human liver cytochrome P-450 content. Midazolam 181-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 3196361-11 1988 Furthermore, and since we demonstrated that midazolam was predominantly transformed by a single cytochrome P-450 enzyme, we could attribute the large inter-individual variations in midazolam metabolism to differences in human liver cytochrome P-450 content. Midazolam 181-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-248 3243660-6 1988 Since many drugs are metabolized by cytochrome P-450, similar to phenazone, it is likely that their elimination in patients with Hodgkin"s disease and in patients with non-Hodgkin"s lymphoma will be also changed. Antipyrine 65-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 2848984-2 1988 The first compound was ketoconazole, a well-known inhibitor not only of fungal cytochrome P-450 but at higher concentrations also of mammalian cytochrome P-450 dependent enzymes. Ketoconazole 23-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 3149823-10 1988 Incubations of granulosa cells with DMBA in the absence or presence of gonadotropins, testosterone or anti--hCG were performed to investigate possible hormonal requirements for the cytochrome P-450 system(s) which metabolize DMBA. 9,10-Dimethyl-1,2-benzanthracene 36-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 3149823-10 1988 Incubations of granulosa cells with DMBA in the absence or presence of gonadotropins, testosterone or anti--hCG were performed to investigate possible hormonal requirements for the cytochrome P-450 system(s) which metabolize DMBA. 9,10-Dimethyl-1,2-benzanthracene 225-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 3191198-0 1988 [Mechanism of electron transport in the cholesterol-hydroxylating system of adrenal cortex mitochondria: a triple complex of adrenodoxin reductase, adrenodoxin and cytochrome P-450]. Cholesterol 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-181 3149823-14 1988 DMBA mono-oxygenase activity in cell cultures was inhibited about 95% by alpha-naphthoflavone (ANF), an inhibitor of certain cytochrome P-450-catalysed activities. 9,10-Dimethyl-1,2-benzanthracene 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 3149823-14 1988 DMBA mono-oxygenase activity in cell cultures was inhibited about 95% by alpha-naphthoflavone (ANF), an inhibitor of certain cytochrome P-450-catalysed activities. alpha-naphthoflavone 73-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 3149823-14 1988 DMBA mono-oxygenase activity in cell cultures was inhibited about 95% by alpha-naphthoflavone (ANF), an inhibitor of certain cytochrome P-450-catalysed activities. alpha-naphthoflavone 95-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 3196394-0 1988 Differentiation of cytochrome P-450 activities with scoparone as substrate. scoparone 52-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 3196394-2 1988 A direct and highly sensitive high performance liquid chromatography (HPLC) method for measuring cytochrome P-450 activities in biological probes, with the substrate scoparone, is described. scoparone 166-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 3044903-0 1988 Disruption of hepatic heme biosynthesis after interaction of xenobiotics with cytochrome P-450. Heme 22-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 3044903-2 1988 Porphyrinogenic chemicals deplete the regulatory free heme pool by interacting with cytochrome P-450 thereby inhibiting heme biosynthesis and/or causing heme breakdown. Heme 54-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 3044903-2 1988 Porphyrinogenic chemicals deplete the regulatory free heme pool by interacting with cytochrome P-450 thereby inhibiting heme biosynthesis and/or causing heme breakdown. Heme 120-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 3044903-2 1988 Porphyrinogenic chemicals deplete the regulatory free heme pool by interacting with cytochrome P-450 thereby inhibiting heme biosynthesis and/or causing heme breakdown. Heme 120-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 3044903-5 1988 Heterocyclic compounds of this type cause mechanism-based inactivation of cytochrome P-450, leading to the formation of N-alkylporphyrins, with ferrochelatase-inhibitory activity resulting in lowering the free heme pool. n-alkylporphyrins 120-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 3044903-5 1988 Heterocyclic compounds of this type cause mechanism-based inactivation of cytochrome P-450, leading to the formation of N-alkylporphyrins, with ferrochelatase-inhibitory activity resulting in lowering the free heme pool. Heme 210-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 3044903-8 1988 In the process, the heme moiety of cytochrome P-450 is destroyed and the free heme pool is lowered. Heme 20-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 3044903-8 1988 In the process, the heme moiety of cytochrome P-450 is destroyed and the free heme pool is lowered. Heme 78-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 3140691-3 1988 The role of a specific cytochrome P-450 form, debrisoquine 4-hydroxylase, in the metabolism of alfentanil was investigated by competitive inhibition experiments over the concentration range 4-100 microM. Alfentanil 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 3196692-1 1988 A cytochrome P-450 (P-450) multigene family codes for several related human liver enzymes, including the P-450 responsible for (S)-mephenytoin 4"-hydroxylation. Mephenytoin 127-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-18 3419164-0 1988 The reoxidation of cytochrome P-450 by paraquat inhibits aldosterone biosynthesis from 18-hydroxycorticosterone. Paraquat 39-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 3419164-0 1988 The reoxidation of cytochrome P-450 by paraquat inhibits aldosterone biosynthesis from 18-hydroxycorticosterone. Aldosterone 57-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 3419164-0 1988 The reoxidation of cytochrome P-450 by paraquat inhibits aldosterone biosynthesis from 18-hydroxycorticosterone. 18-Hydroxycorticosterone 87-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 3419164-1 1988 Paraquat is an artificial electron carrier that captures electrons from reduced cytochrome P-450 instead of the natural acceptors, thus decreasing the concentration of reduced mitochondrial cytochrome P-450. Paraquat 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 3419164-1 1988 Paraquat is an artificial electron carrier that captures electrons from reduced cytochrome P-450 instead of the natural acceptors, thus decreasing the concentration of reduced mitochondrial cytochrome P-450. Paraquat 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-206 3419164-5 1988 In our conditions, the inhibitory role of paraquat seems restricted to a capture of electrons from reduced cytochrome P-450. Paraquat 42-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-123 3419164-6 1988 Under the same conditions metopirone and spironolactone, known to bind cytochrome P-450 11 beta at the steroid binding site, also inhibited the reaction. Metyrapone 26-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 3419164-6 1988 Under the same conditions metopirone and spironolactone, known to bind cytochrome P-450 11 beta at the steroid binding site, also inhibited the reaction. Spironolactone 41-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 3419164-6 1988 Under the same conditions metopirone and spironolactone, known to bind cytochrome P-450 11 beta at the steroid binding site, also inhibited the reaction. Steroids 103-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 3419164-7 1988 Altogether these results show that for aldosterone synthesis from 18-hydroxycorticosterone to take place, the steroid binding site on cytochrome P-450 must be accessible to 18-hydroxycorticosterone and that the cytochrome P-450 must be the direct donor of reducing equivalents. Aldosterone 39-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 3419164-7 1988 Altogether these results show that for aldosterone synthesis from 18-hydroxycorticosterone to take place, the steroid binding site on cytochrome P-450 must be accessible to 18-hydroxycorticosterone and that the cytochrome P-450 must be the direct donor of reducing equivalents. Aldosterone 39-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-227 3419164-7 1988 Altogether these results show that for aldosterone synthesis from 18-hydroxycorticosterone to take place, the steroid binding site on cytochrome P-450 must be accessible to 18-hydroxycorticosterone and that the cytochrome P-450 must be the direct donor of reducing equivalents. 18-Hydroxycorticosterone 66-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 3419164-7 1988 Altogether these results show that for aldosterone synthesis from 18-hydroxycorticosterone to take place, the steroid binding site on cytochrome P-450 must be accessible to 18-hydroxycorticosterone and that the cytochrome P-450 must be the direct donor of reducing equivalents. 18-Hydroxycorticosterone 66-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-227 3419164-7 1988 Altogether these results show that for aldosterone synthesis from 18-hydroxycorticosterone to take place, the steroid binding site on cytochrome P-450 must be accessible to 18-hydroxycorticosterone and that the cytochrome P-450 must be the direct donor of reducing equivalents. Steroids 110-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 3419164-7 1988 Altogether these results show that for aldosterone synthesis from 18-hydroxycorticosterone to take place, the steroid binding site on cytochrome P-450 must be accessible to 18-hydroxycorticosterone and that the cytochrome P-450 must be the direct donor of reducing equivalents. Steroids 110-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-227 3419164-7 1988 Altogether these results show that for aldosterone synthesis from 18-hydroxycorticosterone to take place, the steroid binding site on cytochrome P-450 must be accessible to 18-hydroxycorticosterone and that the cytochrome P-450 must be the direct donor of reducing equivalents. 18-Hydroxycorticosterone 173-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 3419164-8 1988 Hence, cytochrome P-450 appears as the final linking point between 18-hydroxycorticosterone and the reducing equivalents provided by NADPH. 18-Hydroxycorticosterone 67-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 3419164-8 1988 Hence, cytochrome P-450 appears as the final linking point between 18-hydroxycorticosterone and the reducing equivalents provided by NADPH. NADP 133-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 3145044-0 1988 Modulation of hepatic cytochrome P-450 and DT-diaphorase by oral and sub-cutaneous administration of the pro-oxidant fungicide dichlone (2,3-dichloro-1,4-naphthoquinone). dichlone 127-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 3145044-0 1988 Modulation of hepatic cytochrome P-450 and DT-diaphorase by oral and sub-cutaneous administration of the pro-oxidant fungicide dichlone (2,3-dichloro-1,4-naphthoquinone). dichlone 137-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 3404442-7 1988 Imidazole was used because it is a potent inducer of cytochrome P-450 isozyme 3a which is also induced by ethanol. imidazole 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 3404442-7 1988 Imidazole was used because it is a potent inducer of cytochrome P-450 isozyme 3a which is also induced by ethanol. Ethanol 106-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 3404442-8 1988 The cytochrome P-450 in microsomes from a single human subject metabolized halothane at a rate comparable to that found in microsomes from phenobarbital- and imidazole-pretreated rabbits. Halothane 75-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 3404442-11 1988 Antibodies to cytochrome P-450 3a inhibited halothane metabolism by 90% in microsomes from imidazole-pretreated rabbits, suggesting that isozyme 3a catalyzes halothane metabolism in imidazole-pretreated rabbits. Halothane 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 3404442-11 1988 Antibodies to cytochrome P-450 3a inhibited halothane metabolism by 90% in microsomes from imidazole-pretreated rabbits, suggesting that isozyme 3a catalyzes halothane metabolism in imidazole-pretreated rabbits. imidazole 91-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 3404442-11 1988 Antibodies to cytochrome P-450 3a inhibited halothane metabolism by 90% in microsomes from imidazole-pretreated rabbits, suggesting that isozyme 3a catalyzes halothane metabolism in imidazole-pretreated rabbits. Halothane 158-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 3404442-11 1988 Antibodies to cytochrome P-450 3a inhibited halothane metabolism by 90% in microsomes from imidazole-pretreated rabbits, suggesting that isozyme 3a catalyzes halothane metabolism in imidazole-pretreated rabbits. imidazole 182-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 3372517-0 1988 Cytochrome P-450-catalyzed dehydrogenation of 1,4-dihydropyridines. 1,4-dihydropyridine 46-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3067018-3 1988 Metabolic pathways for various drugs have been correlated with debrisoquin metabolism, the observed polymorphism being attributed to variants of cytochrome P-450. Debrisoquin 63-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-161 3263118-3 1988 The olfactory epithelium of male hamsters has been found to be extremely active in the cumene hydroperoxide-supported oxidation of tetramethylphenylenediamine, and this peroxidase activity has been shown to be cytochrome P-450-dependent. cumene hydroperoxide 87-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 210-226 3263118-3 1988 The olfactory epithelium of male hamsters has been found to be extremely active in the cumene hydroperoxide-supported oxidation of tetramethylphenylenediamine, and this peroxidase activity has been shown to be cytochrome P-450-dependent. Tetramethylphenylenediamine 131-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 210-226 3263118-14 1988 It is suggested that this behaviour is due to 4-ethyl DDC acting not only as a suicidal inhibitor but also as an inducer of certain forms of cytochrome P-450 in the liver; in the olfactory epithelium, however, inactivation, but not induction, occurs. 4-ethyl ddc 46-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-157 2839175-2 1988 Lipid peroxidation, monitored by the formation of thiobarbituric acid-reactive substance, was increased with increasing concentration of detergent-solubilized NADPH cytochrome P-450 reductase, cytochrome P-450 or Fe3+-ADP. thiobarbituric acid 50-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-181 2839175-3 1988 Cytochrome P-450-dependent lipid peroxidation was parallel to O-2 generation monitored by chemiluminescence probe with 2-methyl-6-(p-methoxyphenol)-3,7-dihydroimidazo[1,2-a]pyrazin++ +-3-one. 2-methyl-6-(p-methoxyphenol)-3,7-dihydroimidazo[1,2-a]pyrazin++ +-3-one 119-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3203057-15 1988 Although the competitive inhibition between some sulphonamides and tolbutamide is consistent with metabolism by the same isozyme of cytochrome P-450 it does not prove it and further studies with purified enzymes will be necessary to confirm this. Sulfonamides 49-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 3203057-15 1988 Although the competitive inhibition between some sulphonamides and tolbutamide is consistent with metabolism by the same isozyme of cytochrome P-450 it does not prove it and further studies with purified enzymes will be necessary to confirm this. Tolbutamide 67-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 2839175-1 1988 In order to evaluate the O-2 participation in NADPH-dependent microsomal lipid peroxidation, we used reconstructed system which contained detergent-solubilized NADPH-dependent cytochrome P-450 reductase, cytochrome P-450, phospholipid liposomes, NADPH and Fe3+-ADP. NADP 160-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-192 3372517-1 1988 A variety of different 4-substituted 1,4-dihydropyridine Hantzsch esters are substrates for ring dehydrogenation by a cytochrome P-450 (P-450) enzyme (P-450 UT-A); the substitutent could be varied from a hydrogen to a naphthalenyl, but a pyrenyl derivative was not dehydrogenated. 4-substituted 1,4-dihydropyridine hantzsch esters 23-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 3372517-1 1988 A variety of different 4-substituted 1,4-dihydropyridine Hantzsch esters are substrates for ring dehydrogenation by a cytochrome P-450 (P-450) enzyme (P-450 UT-A); the substitutent could be varied from a hydrogen to a naphthalenyl, but a pyrenyl derivative was not dehydrogenated. Hydrogen 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 3372517-1 1988 A variety of different 4-substituted 1,4-dihydropyridine Hantzsch esters are substrates for ring dehydrogenation by a cytochrome P-450 (P-450) enzyme (P-450 UT-A); the substitutent could be varied from a hydrogen to a naphthalenyl, but a pyrenyl derivative was not dehydrogenated. naphthalenyl 218-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 3372517-1 1988 A variety of different 4-substituted 1,4-dihydropyridine Hantzsch esters are substrates for ring dehydrogenation by a cytochrome P-450 (P-450) enzyme (P-450 UT-A); the substitutent could be varied from a hydrogen to a naphthalenyl, but a pyrenyl derivative was not dehydrogenated. pyrenyl 238-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 3378384-5 1988 The data suggest that this cytochrome P-450 is the major cyclosporine-metabolizing enzyme in human liver. Cyclosporine 57-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 3378384-6 1988 The substrate specificity and the known inducers and inhibitors of this cytochrome P-450 explain several clinically observed drug interactions with cyclosporine. Cyclosporine 148-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 3385648-3 1988 An improved method is described in which toxic APAP metabolites are generated by a purified and reconstituted cytochrome P-450 system, minimizing the amount of exogenous detoxification enzymes in the assay. Acetaminophen 47-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 3135288-6 1988 Both cytochrome P-450 (P less than .01) and nicotinamide adenine dinucleotide phosphate (NADPH) cytochrome c reductase (P less than .06) were higher in ewes receiving PB than in controls. Phenobarbital 167-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 3385648-8 1988 The data are consistent with the generation of cytochrome P-450-dependent reactive metabolites which subsequently can be detoxified by glutathione. Glutathione 135-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 3382731-0 1988 [Effects of hydrogen peroxide on cytochrome P-450 inactivation]. Hydrogen Peroxide 12-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 3181604-2 1988 In man and animals, there is an associated rise in microsomal cytochrome P-450, including a specific form (P-450IIEI) with high affinity for ethanol and for the activation of some drugs (i.e. acetaminophen), carcinogens (i.e. N-nitrosodimethylamine) and hepatotoxic agents (i.e. CCl4), thereby contributing to the susceptibility of alcoholics to xenobiotics, including industrial solvents. Ethanol 141-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3181604-2 1988 In man and animals, there is an associated rise in microsomal cytochrome P-450, including a specific form (P-450IIEI) with high affinity for ethanol and for the activation of some drugs (i.e. acetaminophen), carcinogens (i.e. N-nitrosodimethylamine) and hepatotoxic agents (i.e. CCl4), thereby contributing to the susceptibility of alcoholics to xenobiotics, including industrial solvents. Acetaminophen 192-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3181604-2 1988 In man and animals, there is an associated rise in microsomal cytochrome P-450, including a specific form (P-450IIEI) with high affinity for ethanol and for the activation of some drugs (i.e. acetaminophen), carcinogens (i.e. N-nitrosodimethylamine) and hepatotoxic agents (i.e. CCl4), thereby contributing to the susceptibility of alcoholics to xenobiotics, including industrial solvents. Dimethylnitrosamine 226-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3181604-3 1988 In addition, a benzoflavone-inducible liver cytochrome P-450 isoenzyme distinct but catalytically similar to cytochrome P-450IIE1 was purified which may play a significant role in drinkers who also are heavy smokers. Benzoflavones 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-60 3382731-1 1988 Inactivation rate of purified oligomeric cytochrome P-450 LM2 has been investigated in glucose oxidase system and under the action of exogenous hydrogen peroxide (400 microM). Hydrogen Peroxide 144-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 3288213-2 1988 Anti-human NADPH-cytochrome P-450 reductase inhibited all theophylline metabolic pathways confirming the involvement of cytochrome P-450 isozymes in the metabolism of theophylline. Theophylline 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 3382731-2 1988 It has been found that hydrogen peroxide has a distinct inactivating effect on cytochrome P-450. Hydrogen Peroxide 23-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 3288213-2 1988 Anti-human NADPH-cytochrome P-450 reductase inhibited all theophylline metabolic pathways confirming the involvement of cytochrome P-450 isozymes in the metabolism of theophylline. Theophylline 167-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 3288213-12 1988 The results confirm that there are at least two distinct cytochrome P-450 isozymes involved in theophylline metabolism, one isozyme being involved with the demethylations and a different isozyme involved in the 8-hydroxylation pathway. Theophylline 95-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3382731-4 1988 Benzphetamine, a substrate specific for this enzyme isoform, exerts a protective effect by decreasing the rate of cytochrome P-450 inactivation and SH-group oxidation. Benzphetamine 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 3045791-7 1988 Serum LDL-C levels are inversely proportional to hepatic cytochrome P-450 concentrations. ldl-c 6-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3285175-8 1988 Additional experiments did not show any evidence for the involvement of a number of other human cytochrome P-450 enzymes in 17 alpha-ethynylestradiol 2-hydroxylation (i.e., P-450DB, P-450PA, P-450MP, P-450j). Ethinyl Estradiol 124-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 3367901-0 1988 Modulation of rabbit and human hepatic cytochrome P-450-catalyzed steroid hydroxylations by alpha-naphthoflavone. Steroids 66-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 3285175-0 1988 Oxidation of 17 alpha-ethynylestradiol by human liver cytochrome P-450. Ethinyl Estradiol 13-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 3285175-7 1988 In addition, 17 alpha-ethynylestradiol appears to be a mechanism-based inhibitor in human liver microsomes, as shown by the loss of both spectrally detectable cytochrome P-450 and 17 alpha-ethynylestradiol 2-hydroxylase activity during incubation in the presence of NADPH. Ethinyl Estradiol 13-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-175 2898328-1 1988 Halothane is reduced to 2-chloro-1,1,1-trifluoroethane (CTE) and 2-chloro-1,1-difluoroethene (CDE) by cytochrome P-450. Halothane 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 3044468-15 1988 Subjects with a deficiency in a particular isoenzyme of cytochrome P-450 poorly metabolize perhexiline and are at higher risk of developing liver lesions. Perhexiline 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 2899931-4 1988 Histamine and three of these drugs, namely cimetidine, ranitidine and famotidine, were weak inhibitors of this cytochrome P-450-catalysed O-deethylation, but mifentidine was a potent competitive inhibitor with a Ki in the range 40-70 microM. Histamine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 2899931-4 1988 Histamine and three of these drugs, namely cimetidine, ranitidine and famotidine, were weak inhibitors of this cytochrome P-450-catalysed O-deethylation, but mifentidine was a potent competitive inhibitor with a Ki in the range 40-70 microM. Cimetidine 43-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 2899931-4 1988 Histamine and three of these drugs, namely cimetidine, ranitidine and famotidine, were weak inhibitors of this cytochrome P-450-catalysed O-deethylation, but mifentidine was a potent competitive inhibitor with a Ki in the range 40-70 microM. Ranitidine 55-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 2899931-4 1988 Histamine and three of these drugs, namely cimetidine, ranitidine and famotidine, were weak inhibitors of this cytochrome P-450-catalysed O-deethylation, but mifentidine was a potent competitive inhibitor with a Ki in the range 40-70 microM. Famotidine 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 2899931-4 1988 Histamine and three of these drugs, namely cimetidine, ranitidine and famotidine, were weak inhibitors of this cytochrome P-450-catalysed O-deethylation, but mifentidine was a potent competitive inhibitor with a Ki in the range 40-70 microM. mifentidine 158-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 2899931-8 1988 The demonstration that cimetidine, ranitidine and histamine were all poor inhibitors of phenacetin oxidation further suggests the possible lack of identity between the human liver cytochrome P-450 isoenzymes responsible for catalyzing the oxidation of metoprolol and phenacetin. Metoprolol 252-262 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 2899931-8 1988 The demonstration that cimetidine, ranitidine and histamine were all poor inhibitors of phenacetin oxidation further suggests the possible lack of identity between the human liver cytochrome P-450 isoenzymes responsible for catalyzing the oxidation of metoprolol and phenacetin. Phenacetin 267-277 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 3355588-3 1988 Anti-human liver NADPH-cytochrome P-450 reductase IgG inhibited hydroxytolbutamide formation and this metabolite was not formed when NADPH-generating system was omitted from microsomal incubations. hydroxymethyltolbutamide 64-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 3355588-3 1988 Anti-human liver NADPH-cytochrome P-450 reductase IgG inhibited hydroxytolbutamide formation and this metabolite was not formed when NADPH-generating system was omitted from microsomal incubations. NADP 17-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 3355588-4 1988 Tolbutamide hydroxylation followed Michaelis-Menten kinetics, consistent with the involvement of a single form of cytochrome P-450 in this reaction. Tolbutamide 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 2898328-1 1988 Halothane is reduced to 2-chloro-1,1,1-trifluoroethane (CTE) and 2-chloro-1,1-difluoroethene (CDE) by cytochrome P-450. 1,1,1-trifluoro-2-chloroethane 56-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 2898328-1 1988 Halothane is reduced to 2-chloro-1,1,1-trifluoroethane (CTE) and 2-chloro-1,1-difluoroethene (CDE) by cytochrome P-450. 2-chloro-1,1-difluoroethylene 65-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 3367901-0 1988 Modulation of rabbit and human hepatic cytochrome P-450-catalyzed steroid hydroxylations by alpha-naphthoflavone. alpha-naphthoflavone 92-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 3420813-1 1988 Content and catalytic activity of cytochrome P-450 were studied using amidopyrine as a substrate of the I type and aniline as a substrate of the II type. Aminopyrine 70-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 3420813-1 1988 Content and catalytic activity of cytochrome P-450 were studied using amidopyrine as a substrate of the I type and aniline as a substrate of the II type. aniline 115-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 3420813-2 1988 In hypoxia content of cytochrome P-450 and metabolism of amidopyrine were increased, while the enzyme content and the substrate metabolism were decreased in hyperoxia. Aminopyrine 57-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 3133648-6 1988 Lauric acid is a substrate known to be metabolized by a renal cytochrome P-450 to 11 and 12-hydroxylated products. lauric acid 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3133648-8 1988 Based on these results we conclude that cytochrome P-450 plays at the most a limited role in human kidney metabolism of sulindac-sulphide. sulindac sulfide 120-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3390478-1 1988 Studies were carried out of temperature quenching of self-fluorescence of cytochrome P-450 in solution and liposomes from natural phosphatidylcholine, dimiristoylphosphatidylcholine, dipalmitoylphosphatidylcholine. dimiristoylphosphatidylcholine 151-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 3390478-1 1988 Studies were carried out of temperature quenching of self-fluorescence of cytochrome P-450 in solution and liposomes from natural phosphatidylcholine, dimiristoylphosphatidylcholine, dipalmitoylphosphatidylcholine. 1,2-Dipalmitoylphosphatidylcholine 183-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 3390478-2 1988 The fluorescence spectrum of cytochrome P-450 is a superposition of triptophane and tyrosine components. triptophane 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 3390478-2 1988 The fluorescence spectrum of cytochrome P-450 is a superposition of triptophane and tyrosine components. Tyrosine 84-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 2898328-1 1988 Halothane is reduced to 2-chloro-1,1,1-trifluoroethane (CTE) and 2-chloro-1,1-difluoroethene (CDE) by cytochrome P-450. 1,1,1-trifluoro-2-chloroethane 24-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 3356413-0 1988 Phenobarbital induction of cytochrome P-450 b,e genes is dependent on protein synthesis. Phenobarbital 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 3127205-2 1988 The reaction required molecular oxygen and NADPH, and was significantly inhibited by carbon monoxide, suggesting that a cytochrome P-450 is involved. Oxygen 32-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 3127205-2 1988 The reaction required molecular oxygen and NADPH, and was significantly inhibited by carbon monoxide, suggesting that a cytochrome P-450 is involved. NADP 43-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 3127205-2 1988 The reaction required molecular oxygen and NADPH, and was significantly inhibited by carbon monoxide, suggesting that a cytochrome P-450 is involved. Carbon Monoxide 85-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 3127205-3 1988 The neutrophil microsomal fraction showed a carbon monoxide difference spectrum with a peak at 450 nm in the presence of NADPH or dithionite, indicating the presence of a cytochrome P-450. Carbon Monoxide 44-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-187 3127205-3 1988 The neutrophil microsomal fraction showed a carbon monoxide difference spectrum with a peak at 450 nm in the presence of NADPH or dithionite, indicating the presence of a cytochrome P-450. NADP 121-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-187 3127205-3 1988 The neutrophil microsomal fraction showed a carbon monoxide difference spectrum with a peak at 450 nm in the presence of NADPH or dithionite, indicating the presence of a cytochrome P-450. Dithionite 130-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-187 3127205-9 1988 These observations provide direct evidence that the oxygen-activating component of the LTB4 omega-hydroxylase system is a cytochrome P-450. Oxygen 52-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-109 3127205-9 1988 These observations provide direct evidence that the oxygen-activating component of the LTB4 omega-hydroxylase system is a cytochrome P-450. Oxygen 52-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 3356413-1 1988 Phenobarbital induces liver cytochrome P-450 b,e proteins mainly by increasing the rate of transcription of these genes. Phenobarbital 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 3127205-14 1988 These characteristics indicate that the isoenzyme of cytochrome P-450 in human neutrophils, LTB4 omega-hydroxylase, is different from the ones reported to be involved in omega-hydroxylation reactions of prostaglandins and fatty acids. Prostaglandins 203-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 3127205-14 1988 These characteristics indicate that the isoenzyme of cytochrome P-450 in human neutrophils, LTB4 omega-hydroxylase, is different from the ones reported to be involved in omega-hydroxylation reactions of prostaglandins and fatty acids. Prostaglandins 203-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-114 3356413-2 1988 The mechanism responsible for the phenobarbital increment in the rate of transcription of cytochrome P-450 b,e genes is unknown. Phenobarbital 34-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 3127205-14 1988 These characteristics indicate that the isoenzyme of cytochrome P-450 in human neutrophils, LTB4 omega-hydroxylase, is different from the ones reported to be involved in omega-hydroxylation reactions of prostaglandins and fatty acids. Fatty Acids 222-233 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 3127205-14 1988 These characteristics indicate that the isoenzyme of cytochrome P-450 in human neutrophils, LTB4 omega-hydroxylase, is different from the ones reported to be involved in omega-hydroxylation reactions of prostaglandins and fatty acids. Fatty Acids 222-233 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-114 3356413-3 1988 The objective of this study was to assess whether active protein synthesis was needed for phenobarbital to induce the liver cytochrome P-450 b,e genes. Phenobarbital 90-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 3356413-7 1988 While phenobarbital increased cytochrome P-450 b,e mRNAs about 12-fold at 3 hr, this induction was abolished by cycloheximide. Phenobarbital 6-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 3356413-8 1988 To define whether the absence of protein synthesis in hepatocytes inhibited the phenobarbital induction of cytochrome P-450 at the transcriptional level, in vitro transcription rates using isolated nuclei were measured. Phenobarbital 80-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-123 3356413-9 1988 After phenobarbital administration, there was about a 20-fold increment in transcriptional rate of cytochrome P-450 b,e genes. Phenobarbital 6-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 3356413-11 1988 It is proposed that either preexisting regulatory proteins or transacting factors dependent on active protein synthesis participate in the regulation of liver cytochrome P-450 b,e gene transcription after phenobarbital. Phenobarbital 205-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-175 3398207-0 1988 [Metabolism of lidocaine by cytochrome P-450]. Lidocaine 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-45 3341769-0 1988 The role of iron chelates on the selectivity of Fenton reagent in hydroxylation, N-demethylation, and sulfoxidation of cimetidine: a novel biomimetic model for the regioselectivity of cytochrome P-450. Iron 12-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-200 3123495-1 1988 A novel human liver cytochrome P-450 isozyme (P-450-AA), which catalyzes arachidonic acid epoxidation, has been purified to electrophoretic homogeneity from human liver. Arachidonic Acid 73-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 3123495-3 1988 Cytochrome P-450-AA appeared homogeneous as judged by the appearance of a single band on sodium dodecyl sulfate-polyacrylamide gel electrophoresis with an estimated molecular weight of 53,100. polyacrylamide 112-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3123495-4 1988 Although cytochrome P-450-AA had a relatively low specific content of 10.8 nmol/mg, it possessed a high activity of arachidonic acid epoxidation. Arachidonic Acid 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 3123495-6 1988 Moreover, the purified cytochrome P-450-AA catalyzed the de-ethylation of 7-ethoxyresorufin at the rate of 2970 pmol/nmol/min, whereas other cytochrome P-450-dependent reactions were carried out at 23-2,000-fold lower rates and ranged between 0.3-130 pmol/nmol/min. ethoxyresorufin 74-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 3341769-0 1988 The role of iron chelates on the selectivity of Fenton reagent in hydroxylation, N-demethylation, and sulfoxidation of cimetidine: a novel biomimetic model for the regioselectivity of cytochrome P-450. Cimetidine 119-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-200 3341769-4 1988 These results indicate that the oxidation of cimetidine with hydrogen peroxide activated by various chelated ferrous ions serves as a biomimetic model for the regioselectivity of multiple forms of cytochrome P-450 in the metabolism of cimetidine. Cimetidine 45-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-213 3341769-4 1988 These results indicate that the oxidation of cimetidine with hydrogen peroxide activated by various chelated ferrous ions serves as a biomimetic model for the regioselectivity of multiple forms of cytochrome P-450 in the metabolism of cimetidine. Hydrogen Peroxide 61-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-213 3341769-4 1988 These results indicate that the oxidation of cimetidine with hydrogen peroxide activated by various chelated ferrous ions serves as a biomimetic model for the regioselectivity of multiple forms of cytochrome P-450 in the metabolism of cimetidine. Cimetidine 235-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-213 3370255-1 1988 The steady-state levels of aerobic and anaerobic reduction of cytochrome b5 by ascorbic acid and the initial rates of cytochrome b5 reduction in the presence of ascorbic acid and of anaerobic cytochrome P-450 reduction in the presence of NADH were used to calculate the rate constants for cytochrome b5 oxidation. NAD 238-242 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-208 3337745-10 1988 These results suggest that the human cytochrome P-450 isozyme that is immunochemically similar and, thus, homologous to rat P-450d plays a major role in the metabolic activation of several procarcinogens examined, and that the activation of AFB1 is catalyzed by another and, possibly, not phenobarbital-inducible form(s) of human cytochrome P-450. Phenobarbital 289-302 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 3337745-10 1988 These results suggest that the human cytochrome P-450 isozyme that is immunochemically similar and, thus, homologous to rat P-450d plays a major role in the metabolic activation of several procarcinogens examined, and that the activation of AFB1 is catalyzed by another and, possibly, not phenobarbital-inducible form(s) of human cytochrome P-450. Phenobarbital 289-302 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 330-346 2827686-1 1988 The mechanism of action of azole antifungal agents is believed to involve inhibition of fungal cytochrome P-450, and, therefore, an investigation of the interaction of these drugs with mammalian cytochrome P-450 systems should provide some indication of their selectivity as antifungal agents. Azoles 27-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-211 3337722-2 1988 These compounds inhibit oxidation and binding of the substrates of cytochrome P-450 (aminopyrine and aniline), inhibition being of a competitive character. Aminopyrine 85-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3346844-2 1988 Ranitidine is thought to spare phase I hepatic metabolism mediated by cytochrome P-450, unlike its counterpart H2-receptor antagonist cimetidine. Ranitidine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 3337722-2 1988 These compounds inhibit oxidation and binding of the substrates of cytochrome P-450 (aminopyrine and aniline), inhibition being of a competitive character. aniline 101-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3342091-0 1988 Species differences in adrenal spironolactone metabolism: relationship to cytochrome P-450 destruction. Spironolactone 31-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 3350119-3 1988 Its mode of inhibitory action in ecdysone biosynthesis is probably inactivation of cytochrome P-450. Ecdysone 33-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 2827463-3 1988 In the present paper we report the isolation and sequence of a cDNA clone for the human hepatic cytochrome P-450 responsible for mephenytoin (an anticonvulsant) oxidation. Mephenytoin 129-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 3250508-0 1988 Cytochrome P-450 model systems: alkene oxidation. Alkenes 32-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2850750-7 1988 The choice of itraconazole--with its improved tissue affinity, its lower therapeutic dose requirements, and its increased selectivity for fungal cytochrome P-450--demonstrates very well that the use of animal and human pharmacology helps in the design of an antifungal drug with as much effect as possible on the fungus and as little effect as possible on the host. Itraconazole 14-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-161 3276317-3 1988 It would appear that there is greater stereoselective control in the interaction of warfarin with cytochrome P-450 enzymes than that observed for interaction with the receptor, vitamin K1 epoxide reductase. Warfarin 84-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 2854994-0 1988 Radical intermediates in the catalytic cycles of cytochrome P-450. radical 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 2854994-1 1988 Schemes are presented summarizing current knowledge of the mechanism of action of cytochrome P-450 when it functions either as a monooxygenase with molecular oxygen as the oxygen donor or as a peroxygenase with peroxy compounds as the oxygen donor. Oxygen 133-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 3342453-0 1988 Cytochrome P-450-mediated oxidation of substrates by electron-transfer; role of oxygen radicals and of 1- and 2-electron oxidation of paracetamol. Acetaminophen 134-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2854994-1 1988 Schemes are presented summarizing current knowledge of the mechanism of action of cytochrome P-450 when it functions either as a monooxygenase with molecular oxygen as the oxygen donor or as a peroxygenase with peroxy compounds as the oxygen donor. Oxygen 158-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 2854994-1 1988 Schemes are presented summarizing current knowledge of the mechanism of action of cytochrome P-450 when it functions either as a monooxygenase with molecular oxygen as the oxygen donor or as a peroxygenase with peroxy compounds as the oxygen donor. Oxygen 158-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 2854994-3 1988 In addition, cytochrome P-450 catalyzes reductive reactions, including a recently discovered reaction in which organic hydroperoxides are cleaved to yield hydrocarbons and aldehydes or ketones. Hydrogen Peroxide 119-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 2854994-3 1988 In addition, cytochrome P-450 catalyzes reductive reactions, including a recently discovered reaction in which organic hydroperoxides are cleaved to yield hydrocarbons and aldehydes or ketones. Hydrocarbons 155-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 2854994-3 1988 In addition, cytochrome P-450 catalyzes reductive reactions, including a recently discovered reaction in which organic hydroperoxides are cleaved to yield hydrocarbons and aldehydes or ketones. Aldehydes 172-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 2854994-3 1988 In addition, cytochrome P-450 catalyzes reductive reactions, including a recently discovered reaction in which organic hydroperoxides are cleaved to yield hydrocarbons and aldehydes or ketones. Ketones 185-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 2854994-5 1988 Evidence has been obtained that lipid hydroperoxides are physiological substrates for this reductive cleavage reaction catalyzed by cytochrome P-450. Lipid Peroxides 32-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 3132330-2 1988 The enzymatic oxidation of tetrachloro-1,4-hydroquinone (1,4-TCHQ), resulting in covalent binding to protein of tetrachloro-1,4-benzoquinone (1,4-TCBQ), was investigated, with special attention to the involvement of cytochrome P-450 and reactive oxygen species. 2,3,5,6-tetrachlorohydroquinone 27-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-232 3132330-2 1988 The enzymatic oxidation of tetrachloro-1,4-hydroquinone (1,4-TCHQ), resulting in covalent binding to protein of tetrachloro-1,4-benzoquinone (1,4-TCBQ), was investigated, with special attention to the involvement of cytochrome P-450 and reactive oxygen species. 2,3,5,6-tetrachlorohydroquinone 57-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-232 3132330-2 1988 The enzymatic oxidation of tetrachloro-1,4-hydroquinone (1,4-TCHQ), resulting in covalent binding to protein of tetrachloro-1,4-benzoquinone (1,4-TCBQ), was investigated, with special attention to the involvement of cytochrome P-450 and reactive oxygen species. Chloranil 112-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-232 3132330-2 1988 The enzymatic oxidation of tetrachloro-1,4-hydroquinone (1,4-TCHQ), resulting in covalent binding to protein of tetrachloro-1,4-benzoquinone (1,4-TCBQ), was investigated, with special attention to the involvement of cytochrome P-450 and reactive oxygen species. 1,4-tcbq 142-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-232 3132330-2 1988 The enzymatic oxidation of tetrachloro-1,4-hydroquinone (1,4-TCHQ), resulting in covalent binding to protein of tetrachloro-1,4-benzoquinone (1,4-TCBQ), was investigated, with special attention to the involvement of cytochrome P-450 and reactive oxygen species. Reactive Oxygen Species 237-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-232 3132330-7 1988 Inhibition of cytochrome P-450 by metyrapone, which is also known to block the P-450-mediated formation of reactive oxygen species, gave a 80% decrease in binding, while the addition of superoxide dismutase prevented 75% of the covalent binding, almost the same amount as found in anerobic incubations. Metyrapone 34-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 3132330-7 1988 Inhibition of cytochrome P-450 by metyrapone, which is also known to block the P-450-mediated formation of reactive oxygen species, gave a 80% decrease in binding, while the addition of superoxide dismutase prevented 75% of the covalent binding, almost the same amount as found in anerobic incubations. Reactive Oxygen Species 107-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 3132330-8 1988 A large part of the conversion of 1,4-TCHQ to 1,4-TCBQ is apparently not catalyzed by cytochrome P-450 itself, but is mediated by superoxide anion formed by this enzyme. 2,3,5,6-tetrachlorohydroquinone 34-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 3132330-8 1988 A large part of the conversion of 1,4-TCHQ to 1,4-TCBQ is apparently not catalyzed by cytochrome P-450 itself, but is mediated by superoxide anion formed by this enzyme. 1,4-tcbq 46-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 3132330-11 1988 1,4-TCHQ was shown to stimulate the oxidase activity of microsomal cytochrome P-450. 2,3,5,6-tetrachlorohydroquinone 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3288361-0 1988 Cytochrome P450 of fungi: primary target for azole antifungal agents. Azoles 45-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 3288361-9 1988 Cytochrome P450, which mediates the 14 alpha-demethylation of lanosterol, is also present in mammalian cells. Lanosterol 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 3288361-11 1988 If azole antifungal agents inhibit mammalian cytochrome P450 too, their systemic use may result in potentially significant adverse reactions. Azoles 3-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 3276317-4 1988 Indeed, warfarin has been developed as a powerful stereochemical probe for in vitro studies of the terminal enzyme in the mixed-function oxidase system, cytochrome P-450. Warfarin 8-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-169 3243187-3 1988 Our data show also that phenobarbital not only enhances both the direct and metabolism-mediated interaction of most tested PHH with microsomal cytochrome P-450, but also increases the affinity of hexachlorobutadiene, chloroform and carbon tetrachloride for the mitochondrial sites resulting in respiration inhibition. Phenobarbital 24-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 3078287-2 1988 The role of the FMO relative to cytochrome P-450 in the oxidation of the sulfur atoms of organosulfur compounds is considered with particular reference to the hepatatoxicant thiobenzamide, the insecticide phorate and the drug, thioridazine. Sulfur 73-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 2894945-1 1988 Competitive inhibition studies using human liver microsomes have shown that quinidine (QD) has an exceptionally high affinity (60 nM) for the genetically variable cytochrome P-450 that catalyzes the formation of 4-hydroxydebrisoquine and dehydrosparteines from debrisoquine and sparteine. Quinidine 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 2894945-1 1988 Competitive inhibition studies using human liver microsomes have shown that quinidine (QD) has an exceptionally high affinity (60 nM) for the genetically variable cytochrome P-450 that catalyzes the formation of 4-hydroxydebrisoquine and dehydrosparteines from debrisoquine and sparteine. 4-hydroxydebrisoquin 212-233 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 2894945-1 1988 Competitive inhibition studies using human liver microsomes have shown that quinidine (QD) has an exceptionally high affinity (60 nM) for the genetically variable cytochrome P-450 that catalyzes the formation of 4-hydroxydebrisoquine and dehydrosparteines from debrisoquine and sparteine. Debrisoquin 221-233 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 2894945-1 1988 Competitive inhibition studies using human liver microsomes have shown that quinidine (QD) has an exceptionally high affinity (60 nM) for the genetically variable cytochrome P-450 that catalyzes the formation of 4-hydroxydebrisoquine and dehydrosparteines from debrisoquine and sparteine. Sparteine 245-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 2894945-8 1988 These observations indicate that none of the major oxidative reactions of QD are catalyzed by the sparteine/debrisoquine isozyme; QD may simply bind to this cytochrome P-450, without being oxidized by it. Sparteine 98-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 3383988-11 1988 The two main inferences are first, that tolbutamide and antipyrine are metabolised by different forms of cytochrome P-450, and second that a battery of model substrates is needed to investigate the inhibitory effects of a drug in man. Tolbutamide 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 3383988-11 1988 The two main inferences are first, that tolbutamide and antipyrine are metabolised by different forms of cytochrome P-450, and second that a battery of model substrates is needed to investigate the inhibitory effects of a drug in man. Antipyrine 56-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 3128014-13 1988 These data suggest the possibility that propranolol oxidation may be mediated in part, by one or more human liver cytochrome P-450 species catalyzing phenacetin oxidation. Propranolol 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 3406035-0 1988 Role of cytochrome P-450 in hydrocarbon formation from xenobiotic and lipid hydroperoxides. Hydrocarbons 28-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 3406035-0 1988 Role of cytochrome P-450 in hydrocarbon formation from xenobiotic and lipid hydroperoxides. Lipid Peroxides 70-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 3406035-1 1988 Cytochrome P-450 has recently been found to catalyze the reductive cleavage of both xenobiotic hydroperoxides and biologically occurring lipid hydroperoxides. xenobiotic hydroperoxides 84-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3406035-1 1988 Cytochrome P-450 has recently been found to catalyze the reductive cleavage of both xenobiotic hydroperoxides and biologically occurring lipid hydroperoxides. Lipid Peroxides 137-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3406036-0 1988 A unique deuterium/proton exchange during cytochrome P-450 mediated expoxidation of propene and butene. Deuterium 9-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 3406036-0 1988 A unique deuterium/proton exchange during cytochrome P-450 mediated expoxidation of propene and butene. propylene 84-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 3406036-0 1988 A unique deuterium/proton exchange during cytochrome P-450 mediated expoxidation of propene and butene. butylene 96-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 3406036-1 1988 A cytochrome P-450 LM2 reconstituted system mediated expoxidations of small terminal alkenes to the corresponding alkyloxiranes. Alkenes 85-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-18 3406036-1 1988 A cytochrome P-450 LM2 reconstituted system mediated expoxidations of small terminal alkenes to the corresponding alkyloxiranes. alkyloxiranes 114-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-18 3129418-5 1988 The absolute spectrum of the aromatase exhibits a Soret peak at 423 nm in the absence of testosterone and addition of testosterone to the aromatase sample makes its absorption peak shift gradually from 423 to 393 nm (high spin type peak), which is a usual characteristic in the spectrum of cytochrome P-450. Testosterone 118-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 290-306 3128014-13 1988 These data suggest the possibility that propranolol oxidation may be mediated in part, by one or more human liver cytochrome P-450 species catalyzing phenacetin oxidation. Phenacetin 150-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 3689794-1 1987 Consideration of the computer-optimised dimensions of anthraflavic acid indicates that it is essentially a planar molecule with a large area/depth ratio, that would preferentially interact with the polycyclic aromatic hydrocarbon-induced family of cytochrome P-450 proteins (cytochromes P-448). 2,6-dihydroxyanthraquinone 54-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 248-264 3442670-4 1987 We have accumulated evidence that autoimmune antibodies observed in sera of patients with tienilic acid induced hepatitis (anti-liver kidney microsome 2 or anti-LKM2 antibodies) specifically recognize the cytochrome P-450 involved in the mephenytoin hydroxylation polymorphism. Ticrynafen 90-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-221 3442670-4 1987 We have accumulated evidence that autoimmune antibodies observed in sera of patients with tienilic acid induced hepatitis (anti-liver kidney microsome 2 or anti-LKM2 antibodies) specifically recognize the cytochrome P-450 involved in the mephenytoin hydroxylation polymorphism. Mephenytoin 238-249 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-221 3689794-1 1987 Consideration of the computer-optimised dimensions of anthraflavic acid indicates that it is essentially a planar molecule with a large area/depth ratio, that would preferentially interact with the polycyclic aromatic hydrocarbon-induced family of cytochrome P-450 proteins (cytochromes P-448). Polycyclic Aromatic Hydrocarbons 198-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 248-264 3442670-6 1987 The cytochrome P-450 recognized by anti-LKM2 antibodies was immunopurified from microsomes derived from livers of extensive (EM) or poor metabolizers (PM) of (S)-mephenytoin. Mephenytoin 158-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 3435455-5 1987 At higher concentrations of alcohol (25-30 mM), rates of ethanol uptake were about 80 mumol/h per g, whereas rates of butanol uptake were only about 9 mumol/h per g. Because rates of butanol metabolism via cytochrome P-450 in deermice were more than an order of magnitude lower than rates of ethanol uptake in livers from ADH-negative deermice, it is concluded that ethanol uptake by perfused livers from ADH-negative deermice is catalysed predominantly via catalase-H2O2. Ethanol 292-299 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-222 3442670-9 1987 These data strongly suggest that the mephenytoin hydroxylation deficiency is caused by a minor structural change leading to a functionally altered cytochrome P-450 isozyme. Mephenytoin 37-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 3435455-3 1987 Thus butanol is oxidized by cytochrome P-450 in microsomal fractions, but not by cytosolic catalase, in tissues from ADH-negative deermice. Butanols 5-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 3689440-2 1987 The genetically controlled polymorphic oxidation of debrisoquine and sparteine is caused by the absence or functional deficiency of a cytochrome P-450 isozyme. Debrisoquin 52-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 3435455-5 1987 At higher concentrations of alcohol (25-30 mM), rates of ethanol uptake were about 80 mumol/h per g, whereas rates of butanol uptake were only about 9 mumol/h per g. Because rates of butanol metabolism via cytochrome P-450 in deermice were more than an order of magnitude lower than rates of ethanol uptake in livers from ADH-negative deermice, it is concluded that ethanol uptake by perfused livers from ADH-negative deermice is catalysed predominantly via catalase-H2O2. Ethanol 292-299 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-222 3689440-2 1987 The genetically controlled polymorphic oxidation of debrisoquine and sparteine is caused by the absence or functional deficiency of a cytochrome P-450 isozyme. Sparteine 69-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 3435455-5 1987 At higher concentrations of alcohol (25-30 mM), rates of ethanol uptake were about 80 mumol/h per g, whereas rates of butanol uptake were only about 9 mumol/h per g. Because rates of butanol metabolism via cytochrome P-450 in deermice were more than an order of magnitude lower than rates of ethanol uptake in livers from ADH-negative deermice, it is concluded that ethanol uptake by perfused livers from ADH-negative deermice is catalysed predominantly via catalase-H2O2. Alcohols 28-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-222 3689440-3 1987 In order to elucidate the mechanisms underlying the differences in cytochrome P-450 function we have studied the 1"-hydroxylation of the prototype drug bufuralol in human liver microsomes of individuals phenotyped in vivo as extensive metabolizers (EM, N = 10), poor metabolizers (PM, N = 5) and in subjects with an intermediate rate of metabolism (IM, N = 4). bufuralol 152-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3689440-6 1987 Inhibition of bufuralol 1"-hydroxylation by quinidine was biphasic in EM microsomes, providing further support for the involvement of at least two cytochrome P-450 isozymes. bufuralol 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 3689440-8 1987 Our data suggest that the debrisoquine/sparteine type of oxidation polymorphism is caused by an almost complete loss of a minor cytochrome P-450 isozyme which has a high affinity and stereoselectivity for (+)-bufuralol and a high sensitivity to inhibition by quinidine. Debrisoquin 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 3689440-8 1987 Our data suggest that the debrisoquine/sparteine type of oxidation polymorphism is caused by an almost complete loss of a minor cytochrome P-450 isozyme which has a high affinity and stereoselectivity for (+)-bufuralol and a high sensitivity to inhibition by quinidine. Sparteine 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 3689440-8 1987 Our data suggest that the debrisoquine/sparteine type of oxidation polymorphism is caused by an almost complete loss of a minor cytochrome P-450 isozyme which has a high affinity and stereoselectivity for (+)-bufuralol and a high sensitivity to inhibition by quinidine. bufuralol 205-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 3435455-5 1987 At higher concentrations of alcohol (25-30 mM), rates of ethanol uptake were about 80 mumol/h per g, whereas rates of butanol uptake were only about 9 mumol/h per g. Because rates of butanol metabolism via cytochrome P-450 in deermice were more than an order of magnitude lower than rates of ethanol uptake in livers from ADH-negative deermice, it is concluded that ethanol uptake by perfused livers from ADH-negative deermice is catalysed predominantly via catalase-H2O2. Butanols 183-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-222 3435455-5 1987 At higher concentrations of alcohol (25-30 mM), rates of ethanol uptake were about 80 mumol/h per g, whereas rates of butanol uptake were only about 9 mumol/h per g. Because rates of butanol metabolism via cytochrome P-450 in deermice were more than an order of magnitude lower than rates of ethanol uptake in livers from ADH-negative deermice, it is concluded that ethanol uptake by perfused livers from ADH-negative deermice is catalysed predominantly via catalase-H2O2. Hydrogen Peroxide 467-471 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-222 3689440-8 1987 Our data suggest that the debrisoquine/sparteine type of oxidation polymorphism is caused by an almost complete loss of a minor cytochrome P-450 isozyme which has a high affinity and stereoselectivity for (+)-bufuralol and a high sensitivity to inhibition by quinidine. Quinidine 259-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 2834196-8 1987 This increase in enzyme activity was associated with a parallel increase in placental microsomal protein immunochemically related to cytochrome P-450 form 6 [derived from 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced rabbit lung]. Polychlorinated Dibenzodioxins 171-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-149 3690940-10 1987 Other work has shown that inhibition of drug metabolism by methoxsalen is associated with both extensive covalent binding of metabolite(s) of methoxsalen to liver microsomal protein in vitro and in vivo and inactivation of cytochrome P-450. Methoxsalen 59-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-239 2834196-8 1987 This increase in enzyme activity was associated with a parallel increase in placental microsomal protein immunochemically related to cytochrome P-450 form 6 [derived from 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced rabbit lung]. Polychlorinated Dibenzodioxins 208-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-149 2830916-2 1987 DCNQ was shown to be bound to microsomal cytochrome P-450 as a type I substrate; its N-demethylation was catalyzed by cytochrome P-450. dcnq 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 3435446-0 1987 Monoclonal antibodies against human cytochrome P-450 recognizing different pregnenolone 16 alpha-carbonitrile-inducible rat cytochromes P-450. Pregnenolone 75-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 3435446-0 1987 Monoclonal antibodies against human cytochrome P-450 recognizing different pregnenolone 16 alpha-carbonitrile-inducible rat cytochromes P-450. alpha-carbonitrile 91-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 2830916-2 1987 DCNQ was shown to be bound to microsomal cytochrome P-450 as a type I substrate; its N-demethylation was catalyzed by cytochrome P-450. dcnq 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 2830916-3 1987 Cytochrome P-450 and NADPH-cytochrome P-450 reductase are capable of DCNQ reduction to semi- and hydroquinones. dcnq 69-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2830916-3 1987 Cytochrome P-450 and NADPH-cytochrome P-450 reductase are capable of DCNQ reduction to semi- and hydroquinones. Hydroquinones 97-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3445541-2 1987 Phenobarbital-induced cytochrome P-450 was especially sensitive to the influence of all the agents studied, while cytochrome P-450 from untreated animals was the least sensitive; the 3-methyl-cholanthrene-induced cytochrome P-448 exhibited the least sensitivity to guanidine. Phenobarbital 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 2825178-0 1987 12(R)-hydroxyicosatetraenoic acid: a cytochrome-P450-dependent arachidonate metabolite that inhibits Na+,K+-ATPase in the cornea. 12(r)-hydroxyicosatetraenoic acid 0-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 2825178-0 1987 12(R)-hydroxyicosatetraenoic acid: a cytochrome-P450-dependent arachidonate metabolite that inhibits Na+,K+-ATPase in the cornea. Arachidonic Acid 63-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 3445541-2 1987 Phenobarbital-induced cytochrome P-450 was especially sensitive to the influence of all the agents studied, while cytochrome P-450 from untreated animals was the least sensitive; the 3-methyl-cholanthrene-induced cytochrome P-448 exhibited the least sensitivity to guanidine. Methylcholanthrene 183-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 3445541-2 1987 Phenobarbital-induced cytochrome P-450 was especially sensitive to the influence of all the agents studied, while cytochrome P-450 from untreated animals was the least sensitive; the 3-methyl-cholanthrene-induced cytochrome P-448 exhibited the least sensitivity to guanidine. Guanidine 265-274 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 3440754-1 1987 Immunocytochemical studies with a monoclonal antibody (MAb-HL3), which recognises a major isozyme of human hepatic cytochrome P-450, have demonstrated this cytochrome in both cryostat and formalin-fixed paraffin-embedded sections of normal human adult liver. Formaldehyde 188-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 3689370-3 1987 In this study HHR were found to possess cytochrome P-450-dependent 7-ethoxyresorufin-O-deethylase (ERD) activity which measures cytochrome P-450 isoenzymes that are highly specific (in the order of greater than 95%) markers for the metabolic activation of many environmental carcinogenic substances such as the polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 311-343 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3689370-3 1987 In this study HHR were found to possess cytochrome P-450-dependent 7-ethoxyresorufin-O-deethylase (ERD) activity which measures cytochrome P-450 isoenzymes that are highly specific (in the order of greater than 95%) markers for the metabolic activation of many environmental carcinogenic substances such as the polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 311-343 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 3689370-3 1987 In this study HHR were found to possess cytochrome P-450-dependent 7-ethoxyresorufin-O-deethylase (ERD) activity which measures cytochrome P-450 isoenzymes that are highly specific (in the order of greater than 95%) markers for the metabolic activation of many environmental carcinogenic substances such as the polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 345-349 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3689370-3 1987 In this study HHR were found to possess cytochrome P-450-dependent 7-ethoxyresorufin-O-deethylase (ERD) activity which measures cytochrome P-450 isoenzymes that are highly specific (in the order of greater than 95%) markers for the metabolic activation of many environmental carcinogenic substances such as the polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 345-349 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 3675576-1 1987 Cytochrome P-450-ALC, an ethanol-oxidizing form of microsomal cytochrome P-450 (P-450), has been purified from human liver. Ethanol 25-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3631597-2 1987 However, cimetidine is known to inhibit clearance of many drugs by reducing hepatic blood flow and by inhibiting cytochrome P-450 enzymes. Cimetidine 9-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 3665337-6 1987 Based on formation clearance values estimated for 6-, 7-, and 8-hydroxywarfarin, rifampin appears to increase the clearance of the parent drug by induction of the cytochrome P-450 isozyme(s) responsible for aromatic hydroxylation. Rifampin 81-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 3654629-0 1987 P-450 HFLa, a form of cytochrome P-450 purified from human fetal livers, is the 16 alpha-hydroxylase of dehydroepiandrosterone 3-sulfate. Dehydroepiandrosterone Sulfate 104-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 3654629-11 1987 We conclude that P-450 HFLa is a form of cytochrome P-450 involved in the 16 alpha-hydroxylation of DHEA-sulfate. Dehydroepiandrosterone Sulfate 100-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 3440754-1 1987 Immunocytochemical studies with a monoclonal antibody (MAb-HL3), which recognises a major isozyme of human hepatic cytochrome P-450, have demonstrated this cytochrome in both cryostat and formalin-fixed paraffin-embedded sections of normal human adult liver. Paraffin 203-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 3663218-2 1987 Oxidized and dithionite-reduced monomers of cytochrome P-450 were much less thermostable than its initial aggregates, while thermal stability of NADPH-specific flavoprotein did not depend on its aggregation state. Dithionite 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-60 3663218-3 1987 Binding spectra of cytochrome P-450 monomers with benzphetamine were atypical and had an absorbance minimum at 422 nm only. Benzphetamine 50-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 3663218-5 1987 Investigation of the dependence of the initial rates of NADPH-dependent cytochrome P-450 reduction and benzphetamine oxidation on the stoichiometry of the flavoprotein and cytochrome P-450 at their constant total concentration showed that the molar ratio of 1:1 was required for maximal activity. NADP 56-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 3663218-5 1987 Investigation of the dependence of the initial rates of NADPH-dependent cytochrome P-450 reduction and benzphetamine oxidation on the stoichiometry of the flavoprotein and cytochrome P-450 at their constant total concentration showed that the molar ratio of 1:1 was required for maximal activity. NADP 56-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 3663218-5 1987 Investigation of the dependence of the initial rates of NADPH-dependent cytochrome P-450 reduction and benzphetamine oxidation on the stoichiometry of the flavoprotein and cytochrome P-450 at their constant total concentration showed that the molar ratio of 1:1 was required for maximal activity. Benzphetamine 103-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 3663445-14 1987 Overall the in vitro studies are consistent with the in vivo results which suggest the involvement of two cytochrome P-450 isozymes in the metabolism of theophylline. Theophylline 153-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 3040745-1 1987 LTB4 20-hydroxylase (P-450LTB) is the cytochrome P-450 in the microsomes of human polymorphonuclear leukocytes that catalyzes the omega-oxidation of leukotriene B4 (LTB4) to 20-OH LTB4. Leukotriene B4 149-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 3040745-1 1987 LTB4 20-hydroxylase (P-450LTB) is the cytochrome P-450 in the microsomes of human polymorphonuclear leukocytes that catalyzes the omega-oxidation of leukotriene B4 (LTB4) to 20-OH LTB4. Leukotriene B4 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 3040745-1 1987 LTB4 20-hydroxylase (P-450LTB) is the cytochrome P-450 in the microsomes of human polymorphonuclear leukocytes that catalyzes the omega-oxidation of leukotriene B4 (LTB4) to 20-OH LTB4. 20-oh ltb4 174-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 3040745-7 1987 Thus, P-450LTB is a novel cytochrome P-450 of human polymorphonuclear leukocytes with substrate recognition determined by the triene bond configuration and the chirality of the hydroxyl groups. TRIETHYLENETETRAMINE 126-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 3039885-0 1987 Cytochrome P-450-related arachidonic acid metabolites. Arachidonic Acid 25-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3429444-0 1987 Purification of human liver cytochrome P-450 catalyzing testosterone 6 beta-hydroxylation. Testosterone 56-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 3429444-8 1987 These results indicate that P-450 human-1 is a major form of cytochrome P-450 responsible for microsomal testosterone 6 beta-hydroxylation. Testosterone 105-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 3429444-9 1987 Thus, this paper is the first report on human cytochrome P-450 responsible for testosterone 6 beta-hydroxylation, which is the major hydroxylation pathway in human liver microsomes. Testosterone 79-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 3039885-7 1987 Cytochrome P-450-dependent monooxygenases are also present in the lung where, in their suggested capacity as oxygen sensors, they generate metabolites of AA that modify pulmonary vasomotion. Oxygen 31-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3608349-5 1987 The data indicate that a urinary metabolite ratio based on paraxanthine 7-demethylation/8-hydroxylation products reflects systemic caffeine clearance and likely monitors cytochrome P-450 activity inducible by polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 209-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-186 3663765-1 1987 Using the previously obtained data on the substrate-type induction of monooxygenase by xenobiotics of phenobarbital type, the method of conversion of typical substrates for cytochrome P-450 into inducers of biosynthesis of this enzymatic system by blocking in the substrate molecule of the position subjected to oxidative conversion in the enzyme active center was tested. Phenobarbital 102-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-189 3620298-7 1987 These studies suggest that tolbutamide is metabolised by an isozyme of cytochrome P-450 which appears to be distinct from those isozymes metabolising many other drugs. Tolbutamide 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 3608348-5 1987 Therefore rifampin preferentially affects norantipyrine or desmethyl- and 3-hydroxydiazepam metabolic formation, suggesting induction of different (iso)zymes of cytochrome P-450. Rifampin 10-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 3039980-1 1987 Several naphthoquinones, except 2-hydroxy-1,4-naphthoquinone, were found to inhibit microsomal cytochrome P-450-linked monooxygenase activities in rabbit liver and human placenta. Naphthoquinones 8-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 3608349-5 1987 The data indicate that a urinary metabolite ratio based on paraxanthine 7-demethylation/8-hydroxylation products reflects systemic caffeine clearance and likely monitors cytochrome P-450 activity inducible by polycyclic aromatic hydrocarbons. 1,7-dimethylxanthine 59-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-186 3039980-1 1987 Several naphthoquinones, except 2-hydroxy-1,4-naphthoquinone, were found to inhibit microsomal cytochrome P-450-linked monooxygenase activities in rabbit liver and human placenta. lawsone 32-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 3039980-0 1987 Inhibition of cytochrome P-450-linked monooxygenase systems by naphthoquinones. Naphthoquinones 63-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 3039980-4 1987 Spectrophotometric studies revealed that naphthoquinones bind to the cytochrome P-450 component of the monooxygenase complex in both microsomal systems, suggesting that the inhibition is caused by direct interaction of these compounds with the heme. Naphthoquinones 41-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3039980-4 1987 Spectrophotometric studies revealed that naphthoquinones bind to the cytochrome P-450 component of the monooxygenase complex in both microsomal systems, suggesting that the inhibition is caused by direct interaction of these compounds with the heme. Heme 244-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3663627-1 1987 The ferric spin-state equilibrium and relaxation rate of cytochrome P-450 has been examined with temperature jump spectroscopy using a number of camphor analogues known to induce different mixed spin states in the substrate-bound complexes [Gould, P., Gelb, M., & Sligar, S. G. (1981) J. Biol. Camphor 145-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3606650-0 1987 Metabolic activation of the tricyclic antidepressant amineptine by human liver cytochrome P-450. amineptin 53-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 3111480-10 1987 It is concluded that a product or products of propranolol oxidation bind irreversibly but non-selectively to human liver microsomal protein, the enzyme system responsible for the activation of propranolol appears to be related more closely to the cytochrome P-450 system which metabolizes phenacetin than to that metabolising debrisoquine, and radiolabelled propranolol is not a sufficiently specific probe for studying these cytochrome P-450 systems. Propranolol 46-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 247-263 3111480-10 1987 It is concluded that a product or products of propranolol oxidation bind irreversibly but non-selectively to human liver microsomal protein, the enzyme system responsible for the activation of propranolol appears to be related more closely to the cytochrome P-450 system which metabolizes phenacetin than to that metabolising debrisoquine, and radiolabelled propranolol is not a sufficiently specific probe for studying these cytochrome P-450 systems. Propranolol 46-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 426-442 3111480-10 1987 It is concluded that a product or products of propranolol oxidation bind irreversibly but non-selectively to human liver microsomal protein, the enzyme system responsible for the activation of propranolol appears to be related more closely to the cytochrome P-450 system which metabolizes phenacetin than to that metabolising debrisoquine, and radiolabelled propranolol is not a sufficiently specific probe for studying these cytochrome P-450 systems. Propranolol 193-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 247-263 3111480-10 1987 It is concluded that a product or products of propranolol oxidation bind irreversibly but non-selectively to human liver microsomal protein, the enzyme system responsible for the activation of propranolol appears to be related more closely to the cytochrome P-450 system which metabolizes phenacetin than to that metabolising debrisoquine, and radiolabelled propranolol is not a sufficiently specific probe for studying these cytochrome P-450 systems. Propranolol 193-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 426-442 3111480-10 1987 It is concluded that a product or products of propranolol oxidation bind irreversibly but non-selectively to human liver microsomal protein, the enzyme system responsible for the activation of propranolol appears to be related more closely to the cytochrome P-450 system which metabolizes phenacetin than to that metabolising debrisoquine, and radiolabelled propranolol is not a sufficiently specific probe for studying these cytochrome P-450 systems. Phenacetin 289-299 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 247-263 3111480-10 1987 It is concluded that a product or products of propranolol oxidation bind irreversibly but non-selectively to human liver microsomal protein, the enzyme system responsible for the activation of propranolol appears to be related more closely to the cytochrome P-450 system which metabolizes phenacetin than to that metabolising debrisoquine, and radiolabelled propranolol is not a sufficiently specific probe for studying these cytochrome P-450 systems. Debrisoquin 326-338 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 247-263 3111480-10 1987 It is concluded that a product or products of propranolol oxidation bind irreversibly but non-selectively to human liver microsomal protein, the enzyme system responsible for the activation of propranolol appears to be related more closely to the cytochrome P-450 system which metabolizes phenacetin than to that metabolising debrisoquine, and radiolabelled propranolol is not a sufficiently specific probe for studying these cytochrome P-450 systems. Propranolol 193-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 247-263 3111319-6 1987 Arachidonic acid (the precursor of PGE2) may inactivates cytochrome P-450, completely reverses the contractile tension of the DA at both low (4 to 12 torr) and high (511 to 712 torr) oxygen tension and is equally effective in the presence and absence of indomethacin. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3111480-10 1987 It is concluded that a product or products of propranolol oxidation bind irreversibly but non-selectively to human liver microsomal protein, the enzyme system responsible for the activation of propranolol appears to be related more closely to the cytochrome P-450 system which metabolizes phenacetin than to that metabolising debrisoquine, and radiolabelled propranolol is not a sufficiently specific probe for studying these cytochrome P-450 systems. Propranolol 193-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 426-442 3111319-6 1987 Arachidonic acid (the precursor of PGE2) may inactivates cytochrome P-450, completely reverses the contractile tension of the DA at both low (4 to 12 torr) and high (511 to 712 torr) oxygen tension and is equally effective in the presence and absence of indomethacin. Dinoprostone 35-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3111319-6 1987 Arachidonic acid (the precursor of PGE2) may inactivates cytochrome P-450, completely reverses the contractile tension of the DA at both low (4 to 12 torr) and high (511 to 712 torr) oxygen tension and is equally effective in the presence and absence of indomethacin. Oxygen 183-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3111319-6 1987 Arachidonic acid (the precursor of PGE2) may inactivates cytochrome P-450, completely reverses the contractile tension of the DA at both low (4 to 12 torr) and high (511 to 712 torr) oxygen tension and is equally effective in the presence and absence of indomethacin. Indomethacin 254-266 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3111319-9 1987 Metyrapone and phenylimidazole, chemical inhibitors of cytochrome P-450, also relax the ductus. Metyrapone 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 3111319-9 1987 Metyrapone and phenylimidazole, chemical inhibitors of cytochrome P-450, also relax the ductus. phenylimidazole 15-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 3594722-0 1987 Defining the active site of cytochrome P-450: the crystal and molecular structure of an inhibitor, SKF-525A. Proadifen 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 3594722-1 1987 The crystal and molecular structure of the cytochrome P-450 inhibitor, SKF-525A [2-(diethylamino)ethyl 2,2-diphenylpentenoate; proadifen hydrochloride] is described. Proadifen 71-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 3594722-1 1987 The crystal and molecular structure of the cytochrome P-450 inhibitor, SKF-525A [2-(diethylamino)ethyl 2,2-diphenylpentenoate; proadifen hydrochloride] is described. 2-(diethylamino)ethyl 2,2-diphenylpentenoate 81-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 3594722-1 1987 The crystal and molecular structure of the cytochrome P-450 inhibitor, SKF-525A [2-(diethylamino)ethyl 2,2-diphenylpentenoate; proadifen hydrochloride] is described. Proadifen 127-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2891811-0 1987 A comparison of the inhibitory effects of roxatidine acetate hydrochloride and cimetidine on cytochrome P-450-mediated drug-metabolism in mouse hepatic microsomes and in man in vivo. roxatidine acetate 42-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2891811-0 1987 A comparison of the inhibitory effects of roxatidine acetate hydrochloride and cimetidine on cytochrome P-450-mediated drug-metabolism in mouse hepatic microsomes and in man in vivo. Cimetidine 79-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 3306269-5 1987 Theophylline is metabolised within the cytochrome P-450 system, with an average total body clearance of 50 to 60 ml/min. Theophylline 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 3114025-3 1987 The antiserum against the NADPH-cytochrome P-450 reductase component (antiserum denoted RE-DFBIV) gave a single major band at the Mr of the authentic enzyme by immunoblotting after electrophoretic separation of SDS-solubilized microsomes and inhibited both the reductase and aromatase activities in human placental and endometrial microsomes (Tseng, L. and Bellino, F.L. Sodium Dodecyl Sulfate 211-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 3307484-2 1987 The ADH-independent pathway of ethanol metabolism by neural cells appeared to be dependent on one or more isoenzymes of cytochrome P-450. Ethanol 31-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 3036002-10 1987 In the absence of ferric-EDTA (and azide), microsomes catalyzed the oxidation of pyrazole to 4-hydroxypyrazole by a cytochrome P-450-dependent reaction which was independent of hydroxyl radicals. pyrazole 81-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 3036002-10 1987 In the absence of ferric-EDTA (and azide), microsomes catalyzed the oxidation of pyrazole to 4-hydroxypyrazole by a cytochrome P-450-dependent reaction which was independent of hydroxyl radicals. 4-hydroxypyrazole 93-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 3675632-0 1987 [Chemical modification of cysteine residues of cholesterol-hydroxylating cytochrome P-450. Cysteine 26-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 3675632-0 1987 [Chemical modification of cysteine residues of cholesterol-hydroxylating cytochrome P-450. Cholesterol 47-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 3675633-0 1987 [Localization of tetranitromethane-modified tyrosine residues in the polypeptide chain of cholesterol-hydroxylating cytochrome P-450]. Tetranitromethane 17-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-133 3675633-0 1987 [Localization of tetranitromethane-modified tyrosine residues in the polypeptide chain of cholesterol-hydroxylating cytochrome P-450]. Tyrosine 44-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-133 3675633-0 1987 [Localization of tetranitromethane-modified tyrosine residues in the polypeptide chain of cholesterol-hydroxylating cytochrome P-450]. Cholesterol 90-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-133 3675633-3 1987 Tyr-93, -94 are supposedly involved in the active site formation of cytochrome P-450. Tyrosine 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 3109426-1 1987 SKF 525-A (proadifen), a well-known inhibitor of drug metabolism and cytochrome P-450 activity, stimulated the release of prostacyclin (PGI2) from the rabbit aorta in vitro. Proadifen 11-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3632717-1 1987 Immunoblot analysis of liver microsomes from nine patients demonstrated that each contained a cytochrome P-450 that reacted with an antibody directed against the ethanol-inducible rabbit liver cytochrome, P-450 3a. Ethanol 162-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 3632717-6 1987 However, we conclude that there is a cytochrome P-450 present in human liver which is immunochemically and catalytically similar to the ethanol-inducible P-450 of rabbit liver. Ethanol 136-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 3598900-0 1987 Hepatic metabolism of tolbutamide: characterization of the form of cytochrome P-450 involved in methyl hydroxylation and relationship to in vivo disposition. Tolbutamide 22-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3598900-1 1987 In vitro investigations suggest the same human liver cytochrome P-450 that catalyzes S-mephenytoin 4-hydroxylation, P-450MP, is responsible for methyl hydroxylation of the oral hypoglycemic agent tolbutamide. Mephenytoin 85-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 3598900-1 1987 In vitro investigations suggest the same human liver cytochrome P-450 that catalyzes S-mephenytoin 4-hydroxylation, P-450MP, is responsible for methyl hydroxylation of the oral hypoglycemic agent tolbutamide. Tolbutamide 196-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 3628967-2 1987 The incubation of liver microsomes with NADPH and CCl4 under anaerobic conditions (to avoid lipid peroxidation) results in an inhibition of cytochrome P-450 similar to that observed in the in vivo intoxication with a standard dose (0.25 ml/100 g body wt) of CCl4, when the level of covalent binding is 5-10 fold higher than that observed in the in vivo condition. NADP 40-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 3109426-1 1987 SKF 525-A (proadifen), a well-known inhibitor of drug metabolism and cytochrome P-450 activity, stimulated the release of prostacyclin (PGI2) from the rabbit aorta in vitro. Proadifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3579983-10 1987 Since the fast phase of enzymatic lipid peroxidation occurred during the fast phase of destruction of cytochrome P-450, it is postulated that iron made available from cytochrome P-450 is sufficient to promote optimal lipid peroxidation. Iron 142-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 3579320-1 1987 The cytochrome P-450-dependent cholesterol side chain cleavage system of the brain has been studied using nonsynaptic mitochondria as the source of enzymatic activity. Cholesterol 31-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 3579983-10 1987 Since the fast phase of enzymatic lipid peroxidation occurred during the fast phase of destruction of cytochrome P-450, it is postulated that iron made available from cytochrome P-450 is sufficient to promote optimal lipid peroxidation. Iron 142-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-183 3579983-0 1987 NADPH- and linoleic acid hydroperoxide-induced lipid peroxidation and destruction of cytochrome P-450 in hepatic microsomes. NADP 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 3615351-0 1987 Spectral interaction between 4-methylphenol and beta-oestradiol on hepatic microsomal cytochrome P-450: difference between ethanol and phenobarbital pretreatment. beta-oestradiol 48-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 3579983-0 1987 NADPH- and linoleic acid hydroperoxide-induced lipid peroxidation and destruction of cytochrome P-450 in hepatic microsomes. linoleic acid hydroperoxide 11-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 3036162-1 1987 Ketoconazole, an imidazole derivative, is a member of a class of metabolic inhibitors acting specifically at cytochrome-P450 mediated reactions. Ketoconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 3615351-0 1987 Spectral interaction between 4-methylphenol and beta-oestradiol on hepatic microsomal cytochrome P-450: difference between ethanol and phenobarbital pretreatment. 4-cresol 29-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 3036162-1 1987 Ketoconazole, an imidazole derivative, is a member of a class of metabolic inhibitors acting specifically at cytochrome-P450 mediated reactions. imidazole 17-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 3566799-0 1987 Stabilization of the reduced halocarbon-cytochrome P-450 complex of halothane by N-alkanes. Halothane 68-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3593412-2 1987 The nature of the cytochrome P-450-dependent enzyme reactions giving rise to four primary metabolites of caffeine was investigated using microsomes isolated from livers of human kidney donors. Caffeine 105-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 3593412-8 1987 Taken together, the data support suggestions from in vivo studies that a PAH-inducible isozyme of cytochrome P-450 plays a significant role in the biotransformation of caffeine in man. Polycyclic Aromatic Hydrocarbons 73-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 3593412-8 1987 Taken together, the data support suggestions from in vivo studies that a PAH-inducible isozyme of cytochrome P-450 plays a significant role in the biotransformation of caffeine in man. Caffeine 168-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 3566799-0 1987 Stabilization of the reduced halocarbon-cytochrome P-450 complex of halothane by N-alkanes. n-alkanes 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3566799-2 1987 Previous studies have shown that halothane forms such a complex with cytochrome P-450, and the result is a strong absorption at 470 nm. Halothane 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3566799-4 1987 Data are presented which show that several organic solvents (C5-C7N-alkanes) will stabilize the complex formed between halothane and cytochrome P-450. Halothane 119-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-149 3566799-7 1987 Stabilization of the halothane complex in a biological system may facilitate studies to identify precisely the halothane-cytochrome P-450 complex and to clarify the mechanisms of halothane reduction by cytochrome P-450. Halothane 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 3566799-7 1987 Stabilization of the halothane complex in a biological system may facilitate studies to identify precisely the halothane-cytochrome P-450 complex and to clarify the mechanisms of halothane reduction by cytochrome P-450. Halothane 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 3032300-1 1987 Hepatocyte membranes destruction in experimental toxic hepatitis caused by heliotrine administration was accompanied by a 10-fold increase in blood serum activity of aldolase fructose-I-monophosphate, a decrease in cytochrome P-450 content, an increase in the rate of cytochrome P-450 inactivation, as well as a decrease in microsomal glucose-6-phosphatase activity. heliotrine 75-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-231 3032300-1 1987 Hepatocyte membranes destruction in experimental toxic hepatitis caused by heliotrine administration was accompanied by a 10-fold increase in blood serum activity of aldolase fructose-I-monophosphate, a decrease in cytochrome P-450 content, an increase in the rate of cytochrome P-450 inactivation, as well as a decrease in microsomal glucose-6-phosphatase activity. heliotrine 75-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 268-284 3032300-3 1987 Phosphatidylcholine protective action is also manifested in an increase in the activity of glucose-6-phosphatase, a microsomal marker enzyme, up to its control level and in a 20% reduced rate of cytochrome P-450 inactivation. Phosphatidylcholines 0-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-211 3566799-7 1987 Stabilization of the halothane complex in a biological system may facilitate studies to identify precisely the halothane-cytochrome P-450 complex and to clarify the mechanisms of halothane reduction by cytochrome P-450. Halothane 111-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 3566799-7 1987 Stabilization of the halothane complex in a biological system may facilitate studies to identify precisely the halothane-cytochrome P-450 complex and to clarify the mechanisms of halothane reduction by cytochrome P-450. Halothane 111-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 3470755-0 1987 Ferric chloride-catalyzed activation of hydrogen peroxide for the demethylation of N,N-dimethylaniline, the epoxidation of olefins, and the oxidative cleavage of vicinal diols in acetonitrile: a reaction mimic for cytochrome P-450. vicinal diols 162-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-230 3472585-1 1987 In order to determine on which chromosome the gene controlling human cytochrome P-450 (debrisoquine/sparteine type) is located a linkage study of polymorphic sparteine oxidation (PSO) to various polymorphic markers was carried out. Debrisoquin 87-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3472585-1 1987 In order to determine on which chromosome the gene controlling human cytochrome P-450 (debrisoquine/sparteine type) is located a linkage study of polymorphic sparteine oxidation (PSO) to various polymorphic markers was carried out. Sparteine 100-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3472585-1 1987 In order to determine on which chromosome the gene controlling human cytochrome P-450 (debrisoquine/sparteine type) is located a linkage study of polymorphic sparteine oxidation (PSO) to various polymorphic markers was carried out. Sparteine 158-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3472585-4 1987 Thus it can be concluded that the gene controlling human cytochrome P-450 (debrisoquine/sparteine) is situated on the long arm of chromosome 22 in close vicinity to P1. Debrisoquin 75-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3472585-4 1987 Thus it can be concluded that the gene controlling human cytochrome P-450 (debrisoquine/sparteine) is situated on the long arm of chromosome 22 in close vicinity to P1. Sparteine 88-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3470755-0 1987 Ferric chloride-catalyzed activation of hydrogen peroxide for the demethylation of N,N-dimethylaniline, the epoxidation of olefins, and the oxidative cleavage of vicinal diols in acetonitrile: a reaction mimic for cytochrome P-450. ferric chloride 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-230 3470755-0 1987 Ferric chloride-catalyzed activation of hydrogen peroxide for the demethylation of N,N-dimethylaniline, the epoxidation of olefins, and the oxidative cleavage of vicinal diols in acetonitrile: a reaction mimic for cytochrome P-450. Hydrogen Peroxide 40-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-230 3671445-2 1987 From the elimination velocity of these model substances conclusions concerning the activity of 3-methylcholanthrene (caffeine elimination) and phenobarbital inducible isoenzymes (metamizol elimination) of cytochrome P-450 are drawn. Caffeine 117-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-221 3671445-2 1987 From the elimination velocity of these model substances conclusions concerning the activity of 3-methylcholanthrene (caffeine elimination) and phenobarbital inducible isoenzymes (metamizol elimination) of cytochrome P-450 are drawn. Phenobarbital 143-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-221 3671445-2 1987 From the elimination velocity of these model substances conclusions concerning the activity of 3-methylcholanthrene (caffeine elimination) and phenobarbital inducible isoenzymes (metamizol elimination) of cytochrome P-450 are drawn. Dipyrone 179-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-221 3829342-8 1987 We conclude that propafenone is metabolized via the same cytochrome P-450 responsible for debrisoquine"s 4-hydroxylation, and that its pharmacokinetics and concentration-response relationships and the incidence of central nervous system side effects are different in patients of different debrisoquine metabolic phenotype. Propafenone 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3829342-8 1987 We conclude that propafenone is metabolized via the same cytochrome P-450 responsible for debrisoquine"s 4-hydroxylation, and that its pharmacokinetics and concentration-response relationships and the incidence of central nervous system side effects are different in patients of different debrisoquine metabolic phenotype. Debrisoquin 90-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 3470755-0 1987 Ferric chloride-catalyzed activation of hydrogen peroxide for the demethylation of N,N-dimethylaniline, the epoxidation of olefins, and the oxidative cleavage of vicinal diols in acetonitrile: a reaction mimic for cytochrome P-450. acetonitrile 179-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-230 3470755-5 1987 This is in contrast to the prevailing view that cytochrome P-450 acts as a redox catalyst to generate an Fe(V)-oxo species or an Fe(IV)-oxo cation radical as the reactive intermediate. fe(iv)-oxo cation radical 129-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 3494453-0 1987 Inactivation of human liver cytochrome P-450 by the drug methoxsalen and other psoralen derivatives. Methoxsalen 57-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 3104339-6 1987 Purified aromatase cytochrome P-450 displays catalytic characteristics similar to the enzyme in intact microsomes in the aromatization of androstenedione, 19-hydroxyandrostenedione and 19-oxoandrostenedione. Androstenedione 138-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 3104339-6 1987 Purified aromatase cytochrome P-450 displays catalytic characteristics similar to the enzyme in intact microsomes in the aromatization of androstenedione, 19-hydroxyandrostenedione and 19-oxoandrostenedione. 19-hydroxy-4-androstene-3,17-dione 155-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 3104339-6 1987 Purified aromatase cytochrome P-450 displays catalytic characteristics similar to the enzyme in intact microsomes in the aromatization of androstenedione, 19-hydroxyandrostenedione and 19-oxoandrostenedione. 19-oxo-delta(4) androstene-3,17-dione 185-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 3494453-0 1987 Inactivation of human liver cytochrome P-450 by the drug methoxsalen and other psoralen derivatives. Ficusin 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 3494453-1 1987 The effects of psoralen derivatives on cytochrome P-450 have been studied in human liver microsomes. Ficusin 15-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 3494453-2 1987 CO-binding cytochrome P-450 was decreased by 33% after 10 min of incubation with 1.5 mM EDTA, an NADPH-regenerating system and 20 microM methoxsalen (8-methoxypsoralen). Edetic Acid 88-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 3494453-2 1987 CO-binding cytochrome P-450 was decreased by 33% after 10 min of incubation with 1.5 mM EDTA, an NADPH-regenerating system and 20 microM methoxsalen (8-methoxypsoralen). NADP 97-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 3494453-2 1987 CO-binding cytochrome P-450 was decreased by 33% after 10 min of incubation with 1.5 mM EDTA, an NADPH-regenerating system and 20 microM methoxsalen (8-methoxypsoralen). Methoxsalen 137-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 3494453-2 1987 CO-binding cytochrome P-450 was decreased by 33% after 10 min of incubation with 1.5 mM EDTA, an NADPH-regenerating system and 20 microM methoxsalen (8-methoxypsoralen). Methoxsalen 150-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 3494453-9 1987 We conclude that methoxsalen is an extremely potent suicide inhibitor of cytochrome P-450 in human liver microsomes. Methoxsalen 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 3494453-11 1987 In the latter derivative, a methyl group is attached on the furan ring and may hinder its metabolic activation and the inactivation of cytochrome P-450. furan 60-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 3103627-2 1987 Many of these aldehydes have been reported to inhibit hepatic cytochrome P-450. Aldehydes 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3103627-3 1987 Our laboratory has shown that trans,trans-muconaldehyde (a possible metabolite of benzene) as well as acrolein and crotonaldehyde, when added to hepatic microsomes, decreased cytochrome P-450 (measured spectrophotometrically). Hexa-2,4-dienedial 30-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-191 3030331-6 1987 Both the reduction of the quinone imines and the reduction of oxygen were found to be cytochrome P-450 dependent. quinone imines 26-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 3103627-3 1987 Our laboratory has shown that trans,trans-muconaldehyde (a possible metabolite of benzene) as well as acrolein and crotonaldehyde, when added to hepatic microsomes, decreased cytochrome P-450 (measured spectrophotometrically). Benzene 82-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-191 3030331-6 1987 Both the reduction of the quinone imines and the reduction of oxygen were found to be cytochrome P-450 dependent. Oxygen 62-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 3030331-0 1987 Role of hepatic microsomal and purified cytochrome P-450 in one-electron reduction of two quinone imines and concomitant reduction of molecular oxygen. quinone imines 90-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3030331-0 1987 Role of hepatic microsomal and purified cytochrome P-450 in one-electron reduction of two quinone imines and concomitant reduction of molecular oxygen. Oxygen 144-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3103627-3 1987 Our laboratory has shown that trans,trans-muconaldehyde (a possible metabolite of benzene) as well as acrolein and crotonaldehyde, when added to hepatic microsomes, decreased cytochrome P-450 (measured spectrophotometrically). Acrolein 102-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-191 3030331-1 1987 The possible role of cytochrome P-450 in one-electron reduction of quinoid compounds as well as in the formation of reduced oxygen species was investigated in hepatic microsomal and reconstituted systems of purified cytochrome P-450 and purified NADPH-cytochrome P-450 reductase using electron spin resonance (ESR) methods. quinoid 67-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 3030331-4 1987 Both NAPQI and 3,5-dimethyl-NAPQI underwent one-electron reduction in microsomal systems and in fully reconstituted systems of cytochrome P-450 and NADPH-cytochrome P-450 reductase under anaerobic and aerobic conditions. N-acetyl-4-benzoquinoneimine 5-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-143 3030331-7 1987 Both activities of cytochrome P-450 may also be involved in the bioactivation of other compounds with quinoid structural elements, like many chemotherapeutic agents. quinoid 102-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 3103627-3 1987 Our laboratory has shown that trans,trans-muconaldehyde (a possible metabolite of benzene) as well as acrolein and crotonaldehyde, when added to hepatic microsomes, decreased cytochrome P-450 (measured spectrophotometrically). 2-butenal 115-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-191 3030331-4 1987 Both NAPQI and 3,5-dimethyl-NAPQI underwent one-electron reduction in microsomal systems and in fully reconstituted systems of cytochrome P-450 and NADPH-cytochrome P-450 reductase under anaerobic and aerobic conditions. 3,5-Dimethyl-napqi 15-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-143 3816008-6 1987 Erythromycin has known effects on hepatic enzyme function, with altered cytochrome P-450 function. Erythromycin 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 3032244-1 1987 A cDNA sequence related to the human cytochrome P-450 responsible for S-mephenytoin 4-hydroxylation (P-450MP) has been isolated from a human liver bacteriophage lambda gt11 library with antibodies specific for P-450MP. Mephenytoin 70-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 2882985-3 1987 It appears that the high affinity enzyme is a polycyclic aromatic hydrocarbon-inducible isozyme of cytochrome P-450, based on competitive inhibition by 7-ethoxyresorufin and benzo[a]pyrene, and based on a significant (p less than 0.001) correlation between 7-ethoxyresorufin-O-deethylation and methylxanthine demethylation rates. Polycyclic Aromatic Hydrocarbons 46-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 3600619-0 1987 [Electron factors in the properties of cytochrome P-450 and mechanism of activation of molecular oxygen]. Oxygen 97-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 3577206-1 1987 The cytochrome P-450 forms involved in debrisoquine 4-hydroxylation (P-450DB), phenacetin O-deethylation (P-450PA), S-mephenytoin 4-hydroxylation (P-450MP), and nifedipine 1,4-oxidation (P-450NF) have been purified to electrophoretic homogeneity from human liver microsomes. Debrisoquin 39-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 3577206-1 1987 The cytochrome P-450 forms involved in debrisoquine 4-hydroxylation (P-450DB), phenacetin O-deethylation (P-450PA), S-mephenytoin 4-hydroxylation (P-450MP), and nifedipine 1,4-oxidation (P-450NF) have been purified to electrophoretic homogeneity from human liver microsomes. Nifedipine 161-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2882985-3 1987 It appears that the high affinity enzyme is a polycyclic aromatic hydrocarbon-inducible isozyme of cytochrome P-450, based on competitive inhibition by 7-ethoxyresorufin and benzo[a]pyrene, and based on a significant (p less than 0.001) correlation between 7-ethoxyresorufin-O-deethylation and methylxanthine demethylation rates. ethoxyresorufin 152-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 2882985-3 1987 It appears that the high affinity enzyme is a polycyclic aromatic hydrocarbon-inducible isozyme of cytochrome P-450, based on competitive inhibition by 7-ethoxyresorufin and benzo[a]pyrene, and based on a significant (p less than 0.001) correlation between 7-ethoxyresorufin-O-deethylation and methylxanthine demethylation rates. Benzo(a)pyrene 174-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 2882985-3 1987 It appears that the high affinity enzyme is a polycyclic aromatic hydrocarbon-inducible isozyme of cytochrome P-450, based on competitive inhibition by 7-ethoxyresorufin and benzo[a]pyrene, and based on a significant (p less than 0.001) correlation between 7-ethoxyresorufin-O-deethylation and methylxanthine demethylation rates. 7-ethoxyresorufin-o 257-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 2882985-3 1987 It appears that the high affinity enzyme is a polycyclic aromatic hydrocarbon-inducible isozyme of cytochrome P-450, based on competitive inhibition by 7-ethoxyresorufin and benzo[a]pyrene, and based on a significant (p less than 0.001) correlation between 7-ethoxyresorufin-O-deethylation and methylxanthine demethylation rates. methylxanthine 294-308 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 3806608-0 1987 Stereochemical studies on the cytochrome P-450 catalyzed oxidation of (S)-nicotine to the (S)-nicotine delta 1"(5")-iminium species. Nicotine 70-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 3493777-0 1987 Significance of cytochrome P-450 (P-450 HFLa) of human fetal livers in the steroid and drug oxidations. Steroids 75-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 3493777-7 1987 We conclude that P-450 HFLa is a form of cytochrome P-450 which catalyzes testosterone 6 beta-hydroxylation and limited drug oxidations in human fetal and adult livers. Testosterone 74-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 3027095-1 1987 omega-Hydroxylation of leukotriene B4 (LTB4) has been reported in human and rodent polymorphonuclear leukocytes; preliminary information indicates that this metabolism is cytochrome P-450 dependent. Leukotriene B4 23-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-187 3508090-4 1987 Production of the three isomers by microsomal protein occurs only in the presence of NADPH and shows that 2HAA proceeds through a cytochrome P-450 mediated pathway to a reactive intermediate similar to acetaminophen, possibly N-acetyl-o-quinoneimine. 2-hydroxyacetanilide 106-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 3111839-0 1987 Ketoconazole as a possible universal inhibitor of cytochrome P-450 dependent enzymes: its mode of inhibition. Ketoconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 3111839-1 1987 Modes of inhibition and binding of ketoconazole, an orally antimycotic agent, to NADPH-cytochrome P-450 dependent enzymes were investigated using subcellular fractions of human and rat testes, human adrenocortical adenoma tissue and rat adrenals and livers. Ketoconazole 35-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-103 3111839-4 1987 Kinetic studies indicated that ketoconazole bound to cytochrome P-450 and not to other components of monooxygenase systems. Ketoconazole 31-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 3111839-5 1987 Spectrophotometric studies also revealed direct binding of ketoconazole to cytochrome P-450 component by inducing type II difference spectra in all tissue preparations examined, indicating that ketoconazole is possibly a universal inhibitor of NADPH-cytochrome P-450 dependent monooxygenases which are involved in metabolism of many substances including steroids, toxins, carcinogens and others. Ketoconazole 59-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 3111839-5 1987 Spectrophotometric studies also revealed direct binding of ketoconazole to cytochrome P-450 component by inducing type II difference spectra in all tissue preparations examined, indicating that ketoconazole is possibly a universal inhibitor of NADPH-cytochrome P-450 dependent monooxygenases which are involved in metabolism of many substances including steroids, toxins, carcinogens and others. Ketoconazole 59-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 250-266 3111839-5 1987 Spectrophotometric studies also revealed direct binding of ketoconazole to cytochrome P-450 component by inducing type II difference spectra in all tissue preparations examined, indicating that ketoconazole is possibly a universal inhibitor of NADPH-cytochrome P-450 dependent monooxygenases which are involved in metabolism of many substances including steroids, toxins, carcinogens and others. Ketoconazole 194-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 3111839-5 1987 Spectrophotometric studies also revealed direct binding of ketoconazole to cytochrome P-450 component by inducing type II difference spectra in all tissue preparations examined, indicating that ketoconazole is possibly a universal inhibitor of NADPH-cytochrome P-450 dependent monooxygenases which are involved in metabolism of many substances including steroids, toxins, carcinogens and others. Ketoconazole 194-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 250-266 3111839-5 1987 Spectrophotometric studies also revealed direct binding of ketoconazole to cytochrome P-450 component by inducing type II difference spectra in all tissue preparations examined, indicating that ketoconazole is possibly a universal inhibitor of NADPH-cytochrome P-450 dependent monooxygenases which are involved in metabolism of many substances including steroids, toxins, carcinogens and others. Steroids 354-362 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 3032726-6 1987 The rates of cytochrome P-450-dependent FUra formation in the microsomal fraction of liver tissue from two patients (1.1 and 1.3 nmol/mg microsomal protein/30 min) were dramatically reduced to less than half of those of two control subjects (2.4 and 2.7). Fluorouracil 40-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 3806608-0 1987 Stereochemical studies on the cytochrome P-450 catalyzed oxidation of (S)-nicotine to the (S)-nicotine delta 1"(5")-iminium species. (s)-nicotine delta 1"(5")-iminium 90-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 3806608-1 1987 Mammals metabolize the tobacco alkaloid (S)-nicotine primarily to the lactam (S)-cotinine by a pathway involving an initial cytochrome P-450 catalyzed two-electron oxidation at the prochiral 5"-carbon atom. Nicotine 40-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 3806608-1 1987 Mammals metabolize the tobacco alkaloid (S)-nicotine primarily to the lactam (S)-cotinine by a pathway involving an initial cytochrome P-450 catalyzed two-electron oxidation at the prochiral 5"-carbon atom. Lactams 70-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 3806608-1 1987 Mammals metabolize the tobacco alkaloid (S)-nicotine primarily to the lactam (S)-cotinine by a pathway involving an initial cytochrome P-450 catalyzed two-electron oxidation at the prochiral 5"-carbon atom. Cotinine 77-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 3806608-1 1987 Mammals metabolize the tobacco alkaloid (S)-nicotine primarily to the lactam (S)-cotinine by a pathway involving an initial cytochrome P-450 catalyzed two-electron oxidation at the prochiral 5"-carbon atom. Carbon 194-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 3806608-8 1987 These findings suggest that the structure of the complex formed between (S)-nicotine and the active site of cytochrome P-450 is highly ordered and dictates the stereochemical course of the reaction pathway. Nicotine 72-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 2821771-0 1987 Cytochrome P450-dependent arachidonate metabolism in corneal epithelium: formation of biologically active compounds. Arachidonic Acid 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 3573427-0 1987 Action of carbon tetrachloride in the reconstituted monooxygenase system using partially purified cytochrome P-450 and P-448. Carbon Tetrachloride 10-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-124 3621372-0 1987 Quantitative structure-activity relationships in a series of alcohols exhibiting inhibition of cytochrome P-450-mediated aniline hydroxylation. Alcohols 61-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 3304211-9 1987 Mechanistically best known is the program turned on by 3-methylcholanthrene-type inducers which includes enhanced synthesis of certain isoenzymes of cytochrome P-450, GT and probably GSH-transferase. Methylcholanthrene 55-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 3621372-0 1987 Quantitative structure-activity relationships in a series of alcohols exhibiting inhibition of cytochrome P-450-mediated aniline hydroxylation. aniline 121-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 3621370-12 1987 Its likely environmental precursor, CF3Br, which is used as a fire extinguisher and a refrigerant, was found to be reduced by a ferrous porphyrin model for cytochrome P-450 only very slowly and thus is not expected to have a major toxic effect if inhaled. bromotrifluoromethane 36-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 3621372-6 1987 The mode of binding and effect on spin-state equilibria in cytochrome P-450 by alcohols has been elucidated by Testa, whereas an alternative hypothesis based on connectivity correlations has been reported by Sabljic and Sabljic (Mol. Alcohols 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 3621370-12 1987 Its likely environmental precursor, CF3Br, which is used as a fire extinguisher and a refrigerant, was found to be reduced by a ferrous porphyrin model for cytochrome P-450 only very slowly and thus is not expected to have a major toxic effect if inhaled. ferroporphyrin 128-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 3691305-0 1987 [Triphenyldioxan--a new effective inducer of cytochrome P-450]. triphenyldioxan 1-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 3327676-14 1987 The first step in the metabolism involves oxidation to the corresponding pyridine derivative by the cytochrome P-450 system. pyridine 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 3493905-5 1987 The result can be explained by the inability of the antibiotic to form inactive cytochrome P-450 metabolite complexes which can interfere with the metabolism of theophylline. Theophylline 161-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 3609112-3 1987 After pretreatment with cimetidine, which inhibits the activity of cytochrome P-450, the peak plasma concentration and area under the plasma-time concentration curve for nifedipine were increased by a mean 84%. Cimetidine 24-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3609112-3 1987 After pretreatment with cimetidine, which inhibits the activity of cytochrome P-450, the peak plasma concentration and area under the plasma-time concentration curve for nifedipine were increased by a mean 84%. Nifedipine 170-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3111822-1 1987 The commercial mixture of polychlorinated biphenyls (Sovol) studied by biochemical and immunochemical methods was found to be an inducer of a wide range of cytochrome P-450 isoenzymes. Polychlorinated Biphenyls 26-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 3111822-1 1987 The commercial mixture of polychlorinated biphenyls (Sovol) studied by biochemical and immunochemical methods was found to be an inducer of a wide range of cytochrome P-450 isoenzymes. Sovol 53-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 3480222-9 1987 The results indicate that the theophylline-enoxacin interaction may be due to inhibition of the cytochrome P-450 isozymes responsible for theophylline metabolism. Theophylline 30-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 3480222-9 1987 The results indicate that the theophylline-enoxacin interaction may be due to inhibition of the cytochrome P-450 isozymes responsible for theophylline metabolism. Enoxacin 43-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 3480222-9 1987 The results indicate that the theophylline-enoxacin interaction may be due to inhibition of the cytochrome P-450 isozymes responsible for theophylline metabolism. Theophylline 138-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 3496215-1 1987 We estimate the "active" part of cytochrome P-450, which is involved in a special substrate transformation, by measuring the initial change of the production rate as a function of the relaxation transitions between two different steady states of the reaction cycle of cytochrome P-450 using the light-reversibility of the carbon monoxide inhibition. Carbon Monoxide 322-337 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 3496215-1 1987 We estimate the "active" part of cytochrome P-450, which is involved in a special substrate transformation, by measuring the initial change of the production rate as a function of the relaxation transitions between two different steady states of the reaction cycle of cytochrome P-450 using the light-reversibility of the carbon monoxide inhibition. Carbon Monoxide 322-337 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 268-284 3496215-4 1987 An application of our model to the O-deethylation of 7-ethoxycoumarin reveals that--in a time average--10%-15% of the spectroscopically detectable cytochrome P-450 is involved in that transformation. 7-ethoxycoumarin 53-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 3331377-3 1987 Representative examples of the above mechanisms are as following: (1) Ketoconazole possesses inhibitory effects in vitro on cytochrome P-450. Ketoconazole 70-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 3320551-2 1987 The pathway of biosynthesis of these steroids from cholesterol involves a sequence of transformations using cytochrome P-450 enzymes which varies within the adrenal cortex as a result of the differential localization of enzymes within the zones. Steroids 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 3027305-8 1987 Etomidate, ketoconazole, miconazole and metyrapone inhibit cortisol biosynthesis in the human adrenal gland in different manners, which appear to involve the four cytochrome P-450-dependent monooxygenase reactions. Etomidate 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 3027305-8 1987 Etomidate, ketoconazole, miconazole and metyrapone inhibit cortisol biosynthesis in the human adrenal gland in different manners, which appear to involve the four cytochrome P-450-dependent monooxygenase reactions. Ketoconazole 11-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 3027305-8 1987 Etomidate, ketoconazole, miconazole and metyrapone inhibit cortisol biosynthesis in the human adrenal gland in different manners, which appear to involve the four cytochrome P-450-dependent monooxygenase reactions. Miconazole 25-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 2824357-1 1987 The metabolism of N-nitrosodiethylamine (NDEA) and its modulation by inhibitors of cytochrome P450 and prostaglandin H synthetase enzymes was investigated in seven well-differentiated early-passage human lung cancer cell lines. Diethylnitrosamine 18-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-98 2824357-1 1987 The metabolism of N-nitrosodiethylamine (NDEA) and its modulation by inhibitors of cytochrome P450 and prostaglandin H synthetase enzymes was investigated in seven well-differentiated early-passage human lung cancer cell lines. Diethylnitrosamine 41-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-98 3027305-8 1987 Etomidate, ketoconazole, miconazole and metyrapone inhibit cortisol biosynthesis in the human adrenal gland in different manners, which appear to involve the four cytochrome P-450-dependent monooxygenase reactions. Metyrapone 40-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 3320551-2 1987 The pathway of biosynthesis of these steroids from cholesterol involves a sequence of transformations using cytochrome P-450 enzymes which varies within the adrenal cortex as a result of the differential localization of enzymes within the zones. Cholesterol 51-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 3320551-4 1987 These may include the following: (1) the normal morphological and functional zonation of the adrenal cortex may be regulated by gradients of steroids in the adrenal cortex; (2) destruction of cytochrome P-450 enzymes on interaction with certain steroids which act as pseudosubstrates may form part of the pathogenesis of some steroidogenic enzyme deficiencies. Steroids 141-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-208 3320551-4 1987 These may include the following: (1) the normal morphological and functional zonation of the adrenal cortex may be regulated by gradients of steroids in the adrenal cortex; (2) destruction of cytochrome P-450 enzymes on interaction with certain steroids which act as pseudosubstrates may form part of the pathogenesis of some steroidogenic enzyme deficiencies. Steroids 245-253 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-208 3320551-6 1987 Under some pathological conditions, individual cytochrome P-450 enzyme activities may become rate-limiting, with consequent overproduction of precursor steroids, leading to mineralocorticoid or androgen excess. Steroids 152-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 3449750-2 1987 The first proposed step is enzymatic alpha-hydroxylation by the active oxygen species of cytochrome P-450, followed by nonenzymatic N-dealkylation and formation of diazohydroxides (RNNOH). Oxygen 71-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 3449750-2 1987 The first proposed step is enzymatic alpha-hydroxylation by the active oxygen species of cytochrome P-450, followed by nonenzymatic N-dealkylation and formation of diazohydroxides (RNNOH). pyrazine-2-diazohydroxide 164-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 3099185-0 1987 Aflatoxin B1 activation to a plasmid mutagen by a chemical model of cytochrome P-450. Aflatoxin B1 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 3099185-1 1987 Aflatoxin B1 (AFB1) was oxidised by a chemical model of cytochrome P-450 and the products obtained analysed by reversed-phase hplc. Aflatoxin B1 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 3099185-1 1987 Aflatoxin B1 (AFB1) was oxidised by a chemical model of cytochrome P-450 and the products obtained analysed by reversed-phase hplc. Aflatoxin B1 14-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 3449750-8 1987 This reaction appears to be a viable one to formation of an ultimate carcinogen by parent dialkylnitrosamines in the hydrophobic substrate binding site of cytochrome P-450. dialkylnitrosamines 90-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 3628472-0 1987 Mechanisms of phenobarbital-type induction of cytochrome P-450 isozymes. Phenobarbital 14-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 3540968-5 1987 It is also shown that anti-cytochrome P-450-8 antibodies as well as human serum containing anti-LKM2 antibodies specifically inhibit the hydroxylation of tienilic acid by human liver microsomes. Ticrynafen 154-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 2895455-5 1987 The interaction that occurs with theophylline and warfarin when the cytochrome P-450 enzyme system is inhibited by cimetidine and ranitidine requires monitoring. Theophylline 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2895455-5 1987 The interaction that occurs with theophylline and warfarin when the cytochrome P-450 enzyme system is inhibited by cimetidine and ranitidine requires monitoring. Warfarin 50-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2895455-5 1987 The interaction that occurs with theophylline and warfarin when the cytochrome P-450 enzyme system is inhibited by cimetidine and ranitidine requires monitoring. Cimetidine 115-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2895455-5 1987 The interaction that occurs with theophylline and warfarin when the cytochrome P-450 enzyme system is inhibited by cimetidine and ranitidine requires monitoring. Ranitidine 130-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 3540968-6 1987 These results indicate that anti-LKM2 antibodies appearing in patients with hepatitis and concomitant administration of tienilic acid are directed against a cytochrome P-450 isoenzyme that catalyzes the metabolic oxidation of this drug. Ticrynafen 120-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 3030012-10 1987 The interaction with mammalian cytochrome P-450 decreases from miconazole greater than ketoconazole much greater than itraconazole and is much weaker than the interaction of the antimycotics with yeast cytochrome P-450. Miconazole 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 2892251-2 1987 It is well documented that cimetidine can inhibit the hepatic elimination of many drugs by binding to the cytochrome P-450 system. Cimetidine 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 2892251-6 1987 Similarly, binding to cytochrome P-450 of human liver microsomes could be seen only with cimetidine and oxmetidine, whereas nizatidine did not affect the binding spectra. Cimetidine 89-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 2892251-6 1987 Similarly, binding to cytochrome P-450 of human liver microsomes could be seen only with cimetidine and oxmetidine, whereas nizatidine did not affect the binding spectra. oxmetidine 104-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 3810888-4 1987 A model is suggested which shows the phenobarbital participation in the formation of the specific configuration of the active site of cytochrome P-450 synthesized; the latter catalyzes the oxidation of a number of substrates by the way typical of inducer itself. Phenobarbital 37-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 3333881-8 1987 Apart from its clinical and rodenticidal uses, warfarin is an excellent substrate for probing the heterogeneity of cytochrome P.450, since its metabolic oxidation is mediated by this mixed function oxidase. Warfarin 47-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 3030012-10 1987 The interaction with mammalian cytochrome P-450 decreases from miconazole greater than ketoconazole much greater than itraconazole and is much weaker than the interaction of the antimycotics with yeast cytochrome P-450. Miconazole 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 3030012-10 1987 The interaction with mammalian cytochrome P-450 decreases from miconazole greater than ketoconazole much greater than itraconazole and is much weaker than the interaction of the antimycotics with yeast cytochrome P-450. Ketoconazole 87-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 3030012-10 1987 The interaction with mammalian cytochrome P-450 decreases from miconazole greater than ketoconazole much greater than itraconazole and is much weaker than the interaction of the antimycotics with yeast cytochrome P-450. Ketoconazole 87-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 3030012-10 1987 The interaction with mammalian cytochrome P-450 decreases from miconazole greater than ketoconazole much greater than itraconazole and is much weaker than the interaction of the antimycotics with yeast cytochrome P-450. Itraconazole 118-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 3790167-9 1986 Under these conditions, the peroxidatic activity of catalase measured in vitro and the ethanol-dependent decrease in catalase-H2O2 in perfused livers also returned to near basal levels; however, the oxidation of ethanol by cytochrome P-450 was inhibited completely. Ethanol 87-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-239 3782137-2 1986 The cDNAs encoding ethanol-inducible forms of rat and human cytochrome P-450s have been isolated, sequenced, and used to study the expression of this cytochrome P-450 during development and by various inducing agents. Ethanol 19-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 3790167-9 1986 Under these conditions, the peroxidatic activity of catalase measured in vitro and the ethanol-dependent decrease in catalase-H2O2 in perfused livers also returned to near basal levels; however, the oxidation of ethanol by cytochrome P-450 was inhibited completely. Ethanol 212-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-239 3025521-5 1986 Since ketoconazole is known to inhibit cytochrome P-450-dependent enzyme reactions, the results of the present study support the contention that cytochrome P-450 is involved in the aromatisation process. Ketoconazole 6-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 3096378-0 1986 Mephenytoin-type polymorphism of drug oxidation: purification and characterization of a human liver cytochrome P-450 isozyme catalyzing microsomal mephenytoin hydroxylation. Mephenytoin 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 3096378-0 1986 Mephenytoin-type polymorphism of drug oxidation: purification and characterization of a human liver cytochrome P-450 isozyme catalyzing microsomal mephenytoin hydroxylation. Mephenytoin 147-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 3096378-2 1986 By monitoring the activities of the two major oxidative pathways of mephenytoin metabolism in the column eluates, we have purified from human livers a cytochrome P-450 isozyme, P-450 meph, which exclusively and stereoselectively catalyzes the 4-hydroxylation of (S)-mephenytoin, the major pathway affected by the polymorphism, whereas P-450 meph was virtually devoid of catalytic activity for N-demethylation of mephenytoin, the pathway remaining unaffected by the genetic deficiency. Mephenytoin 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 3096378-2 1986 By monitoring the activities of the two major oxidative pathways of mephenytoin metabolism in the column eluates, we have purified from human livers a cytochrome P-450 isozyme, P-450 meph, which exclusively and stereoselectively catalyzes the 4-hydroxylation of (S)-mephenytoin, the major pathway affected by the polymorphism, whereas P-450 meph was virtually devoid of catalytic activity for N-demethylation of mephenytoin, the pathway remaining unaffected by the genetic deficiency. Mephenytoin 262-277 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 3096378-2 1986 By monitoring the activities of the two major oxidative pathways of mephenytoin metabolism in the column eluates, we have purified from human livers a cytochrome P-450 isozyme, P-450 meph, which exclusively and stereoselectively catalyzes the 4-hydroxylation of (S)-mephenytoin, the major pathway affected by the polymorphism, whereas P-450 meph was virtually devoid of catalytic activity for N-demethylation of mephenytoin, the pathway remaining unaffected by the genetic deficiency. Mephenytoin 266-277 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 3025521-5 1986 Since ketoconazole is known to inhibit cytochrome P-450-dependent enzyme reactions, the results of the present study support the contention that cytochrome P-450 is involved in the aromatisation process. Ketoconazole 6-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-161 22283296-0 1986 Oxygen activation by metalloporphyrins related to peroxidase and cytochrome P-450. Oxygen 0-6 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 3021236-1 1986 A mitochondrial preparation from duck adrenal gland was used, under aerobic conditions, to show that the oxygen requirement for the last step of aldosterone biosynthesis (transformation of 18-hydroxycorticosterone into aldosterone) is at the cytochrome P-450 level only. Oxygen 105-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 242-258 3021236-1 1986 A mitochondrial preparation from duck adrenal gland was used, under aerobic conditions, to show that the oxygen requirement for the last step of aldosterone biosynthesis (transformation of 18-hydroxycorticosterone into aldosterone) is at the cytochrome P-450 level only. Aldosterone 145-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 242-258 3021236-1 1986 A mitochondrial preparation from duck adrenal gland was used, under aerobic conditions, to show that the oxygen requirement for the last step of aldosterone biosynthesis (transformation of 18-hydroxycorticosterone into aldosterone) is at the cytochrome P-450 level only. 18-Hydroxycorticosterone 189-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 242-258 3021236-1 1986 A mitochondrial preparation from duck adrenal gland was used, under aerobic conditions, to show that the oxygen requirement for the last step of aldosterone biosynthesis (transformation of 18-hydroxycorticosterone into aldosterone) is at the cytochrome P-450 level only. Aldosterone 219-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 242-258 3021236-2 1986 Vitamin C and tetramethyl-p-phenylene-diamine (TMPD) were used to increase oxygen consumption at the cytochrome a3 level, thereby decreasing its availability to cytochrome P-450. Ascorbic Acid 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 3021236-2 1986 Vitamin C and tetramethyl-p-phenylene-diamine (TMPD) were used to increase oxygen consumption at the cytochrome a3 level, thereby decreasing its availability to cytochrome P-450. tetramethyl-p-phenylenediamine 14-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 3021236-2 1986 Vitamin C and tetramethyl-p-phenylene-diamine (TMPD) were used to increase oxygen consumption at the cytochrome a3 level, thereby decreasing its availability to cytochrome P-450. tetramethyl-p-phenylenediamine 47-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 3021236-2 1986 Vitamin C and tetramethyl-p-phenylene-diamine (TMPD) were used to increase oxygen consumption at the cytochrome a3 level, thereby decreasing its availability to cytochrome P-450. Oxygen 75-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 3021236-8 1986 According to polarographic and electron microscopy studies, the reversal of inhibition can only be explained by an increased availability of oxygen at the cytochrome P-450 level. Oxygen 141-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 3021236-9 1986 Experiments performed under aerobic conditions, without a nitrogen atmosphere, show that oxygen is required in the transformation of 18-hydroxycorticosterone into aldosterone, at the cytochrome P-450 level. Oxygen 89-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 3021236-9 1986 Experiments performed under aerobic conditions, without a nitrogen atmosphere, show that oxygen is required in the transformation of 18-hydroxycorticosterone into aldosterone, at the cytochrome P-450 level. 18-Hydroxycorticosterone 133-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 3021236-9 1986 Experiments performed under aerobic conditions, without a nitrogen atmosphere, show that oxygen is required in the transformation of 18-hydroxycorticosterone into aldosterone, at the cytochrome P-450 level. Aldosterone 163-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 3787626-6 1986 Exposure to DSF alone decreased cytochrome P450 levels, but in combination with DCE, the decrement of cytochrome P450 was additive in a DCE concentration-dependent manner. ethylene dichloride 80-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-117 3787626-6 1986 Exposure to DSF alone decreased cytochrome P450 levels, but in combination with DCE, the decrement of cytochrome P450 was additive in a DCE concentration-dependent manner. ethylene dichloride 136-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-117 3787626-7 1986 However, depression of cytochrome P450 by DCE alone was not concentration dependent. ethylene dichloride 42-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 3022748-0 1986 Differential effects of the cytochrome P-450/reductase ratio on the oxidation of ethanol and the hydroxyl radical scavenging agent 2-keto-4-thiomethylbutyric acid (KMBA). Ethanol 81-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-54 3022748-0 1986 Differential effects of the cytochrome P-450/reductase ratio on the oxidation of ethanol and the hydroxyl radical scavenging agent 2-keto-4-thiomethylbutyric acid (KMBA). Hydroxyl Radical 97-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-54 3022748-0 1986 Differential effects of the cytochrome P-450/reductase ratio on the oxidation of ethanol and the hydroxyl radical scavenging agent 2-keto-4-thiomethylbutyric acid (KMBA). kmba 164-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-54 3022748-2 1986 The effect of various ratios of cytochrome P-450 (phenobarbital-inducible isozyme)/reductase on the oxidation of ethanol and KMBA was determined: There was essentially no increase in KMBA oxidation over the range of ratios from 0.5 to 5 as compared to the reductase-catalyzed rate. Phenobarbital 50-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 3022748-2 1986 The effect of various ratios of cytochrome P-450 (phenobarbital-inducible isozyme)/reductase on the oxidation of ethanol and KMBA was determined: There was essentially no increase in KMBA oxidation over the range of ratios from 0.5 to 5 as compared to the reductase-catalyzed rate. Ethanol 113-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 3464943-1 1986 Human liver cytochrome P-450NF is the form of cytochrome P-450 responsible for the oxidation of the calcium-channel blocker nifedipine, which has been reported to show polymorphism in clinical studies. Nifedipine 124-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 22283296-0 1986 Oxygen activation by metalloporphyrins related to peroxidase and cytochrome P-450. Metalloporphyrins 21-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 3757403-6 1986 These results are consistent with cimetidine inhibiting cytochrome P-450 enzymes in the liver and also competing with triamterene for renal tubular secretion. Cimetidine 34-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 3096315-7 1986 The stereochemical and regiochemical differences between oxidation of BA-3,4-diol by peroxyl radicals and cytochrome P-450 are dramatic and suggest that BA-3,4-diol is uniquely suited as a probe to quantitate peroxyl radical-dependent epoxidation in vitro and in vivo. ba-3,4-diol 153-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 3753522-0 1986 Purification of a desmethylimipramine and debrisoquine hydroxylating cytochrome P-450 from human liver. Debrisoquin 42-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 3745165-1 1986 Purification and characterization of two functionally different human liver cytochrome P-450 isozymes involved in impaired hydroxylation of the prototype substrate bufuralol. bufuralol 164-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 3745165-2 1986 The debrisoquine/sparteine-type polymorphism of drug oxidation presumably is caused by the absence or deficiency of cytochrome P-450 (P-450) isozyme(s). Debrisoquin 4-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 3745165-2 1986 The debrisoquine/sparteine-type polymorphism of drug oxidation presumably is caused by the absence or deficiency of cytochrome P-450 (P-450) isozyme(s). Sparteine 17-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 3489441-4 1986 These results suggest that increased testicular heme oxygenase activity is associated with decreased microsomal heme and cytochrome P-450 content during human chorionic gonadotropin-induced desensitization. Heme 48-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 3746811-6 1986 Oxidation of the 4-alkyl compounds led not only to the loss of P-450NF but also to decreases in catalytic activities of cytochrome P-450 isozymes catalyzing other reactions (phenacetin O-deethylation and hexobarbital 3"-hydroxylation). 4-alkyl 17-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 3746811-6 1986 Oxidation of the 4-alkyl compounds led not only to the loss of P-450NF but also to decreases in catalytic activities of cytochrome P-450 isozymes catalyzing other reactions (phenacetin O-deethylation and hexobarbital 3"-hydroxylation). Phenacetin 174-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 3746811-6 1986 Oxidation of the 4-alkyl compounds led not only to the loss of P-450NF but also to decreases in catalytic activities of cytochrome P-450 isozymes catalyzing other reactions (phenacetin O-deethylation and hexobarbital 3"-hydroxylation). Hexobarbital 204-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 3748010-0 1986 Oxidation of quinidine by human liver cytochrome P-450. Quinidine 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 3481957-1 1986 A phenobarbitone inducible cytochrome P-450 gene family (CYP1) has recently been localized to chromosome 19q13.1-qter. Phenobarbital 2-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 2425812-2 1986 The genotoxic activities of cyclophosphamide and its direct acting metabolite, phosphoramide mustard, were studied in the hepatoma cells as cyclophosphamide is known to be metabolized by phenobarbital-inducible cytochrome P-450-associated metabolic activity. Cyclophosphamide 28-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-227 2425812-2 1986 The genotoxic activities of cyclophosphamide and its direct acting metabolite, phosphoramide mustard, were studied in the hepatoma cells as cyclophosphamide is known to be metabolized by phenobarbital-inducible cytochrome P-450-associated metabolic activity. Cyclophosphamide 140-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-227 2425812-5 1986 The cytochrome P-450-associated enzyme inhibitors, SKF-525A and metyrapone, were found to reduce the level of cyclophosphamide-induced sister chromatid exchanges in HepG2 and H4-II-E, suggesting that cyclophosphamide was activated by this pathway in both hepatoma lines. Metyrapone 64-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2425812-5 1986 The cytochrome P-450-associated enzyme inhibitors, SKF-525A and metyrapone, were found to reduce the level of cyclophosphamide-induced sister chromatid exchanges in HepG2 and H4-II-E, suggesting that cyclophosphamide was activated by this pathway in both hepatoma lines. Cyclophosphamide 110-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2425812-5 1986 The cytochrome P-450-associated enzyme inhibitors, SKF-525A and metyrapone, were found to reduce the level of cyclophosphamide-induced sister chromatid exchanges in HepG2 and H4-II-E, suggesting that cyclophosphamide was activated by this pathway in both hepatoma lines. Cyclophosphamide 200-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2425812-6 1986 Direct evidence for the presence of mRNA transcript coding for a phenobarbital-inducible cytochrome P-450 was demonstrated in HepG2 cells by Northern blot analysis. Phenobarbital 65-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 3729971-8 1986 These findings suggest that the inhibition of aminopyrine N-demethylase activity by primaquine is mediated via an interaction with the oxidised form of cytochrome P-450. Primaquine 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-168 3729971-9 1986 In addition, the apparent change in inhibition kinetics suggests a concentration-dependent change in the capacity of primaquine to modulate substrate binding to cytochrome P-450 as well as the formation of a P-450-aminopyrine-primaquine ternary complex. Primaquine 117-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 3741500-2 1986 The metabolism of this drug to its principal metabolite, 4-hydroxydebrisoquine, is catalyzed by a discrete isozyme of cytochrome P-450. 4-hydroxydebrisoquin 57-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 2425856-0 1986 [Induction of the phenobarbital form of cytochrome P-450 by chemically unrelated compounds]. Phenobarbital 18-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 2425856-1 1986 The induction of the phenobarbital form of cytochrome P-450 by xenobiotics (phenobarbital, PB, hexachlorobenzene, HCB; hexachlorocyclohexane. Phenobarbital 21-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2425856-1 1986 The induction of the phenobarbital form of cytochrome P-450 by xenobiotics (phenobarbital, PB, hexachlorobenzene, HCB; hexachlorocyclohexane. Phenobarbital 76-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2425856-1 1986 The induction of the phenobarbital form of cytochrome P-450 by xenobiotics (phenobarbital, PB, hexachlorobenzene, HCB; hexachlorocyclohexane. Lead 91-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2425856-1 1986 The induction of the phenobarbital form of cytochrome P-450 by xenobiotics (phenobarbital, PB, hexachlorobenzene, HCB; hexachlorocyclohexane. Hexachlorobenzene 95-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2425856-1 1986 The induction of the phenobarbital form of cytochrome P-450 by xenobiotics (phenobarbital, PB, hexachlorobenzene, HCB; hexachlorocyclohexane. Hexachlorobenzene 114-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2425856-1 1986 The induction of the phenobarbital form of cytochrome P-450 by xenobiotics (phenobarbital, PB, hexachlorobenzene, HCB; hexachlorocyclohexane. Hexachlorocyclohexane 119-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2425856-4 1986 Using monospecific antibodies against the cytochrome P-450 form isolated from PB-induced microsomes (PB-cytochrome P-450), a double immunodiffusion test revealed immunological identity of cytochrome P-450 forms induced by phenobarbital and other xenobiotics. Phenobarbital 222-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 2425856-4 1986 Using monospecific antibodies against the cytochrome P-450 form isolated from PB-induced microsomes (PB-cytochrome P-450), a double immunodiffusion test revealed immunological identity of cytochrome P-450 forms induced by phenobarbital and other xenobiotics. Phenobarbital 222-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 2425856-4 1986 Using monospecific antibodies against the cytochrome P-450 form isolated from PB-induced microsomes (PB-cytochrome P-450), a double immunodiffusion test revealed immunological identity of cytochrome P-450 forms induced by phenobarbital and other xenobiotics. Phenobarbital 222-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 2425856-6 1986 Antibodies against PB-cytochrome P-450 inhibited by 50-70% the benzphetamine-N-demethylase and aldrin-epoxidase activities, whereas the antibodies to methylcholanthrene-induced cytochrome P-450 were fairly ineffective. Methylcholanthrene 150-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 2425856-6 1986 Antibodies against PB-cytochrome P-450 inhibited by 50-70% the benzphetamine-N-demethylase and aldrin-epoxidase activities, whereas the antibodies to methylcholanthrene-induced cytochrome P-450 were fairly ineffective. Methylcholanthrene 150-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 3709029-10 1986 Moreover, this ratio may be used to detect drug interactions that involve the cytochrome P-450 isozyme(s) responsible for the polymorphic 4-hydroxylation of mephenytoin. Mephenytoin 157-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 3742644-0 1986 The interaction of a homologous series of hydrocarbons with hepatic cytochrome P-450. Hydrocarbons 42-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 3742644-2 1986 The spectral interaction of a homologous series of alkyl-substituted benzenes and related compounds with purified mammalian cytochrome P-450 has been investigated. alkyl-substituted benzenes 51-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 3742644-3 1986 Each of the 10 hydrocarbons produced a Type I spectral change, indicative of a low to high spin transition of the haem iron of cytochrome P-450. Hydrocarbons 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-143 3742644-3 1986 Each of the 10 hydrocarbons produced a Type I spectral change, indicative of a low to high spin transition of the haem iron of cytochrome P-450. Iron 119-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-143 3742644-9 1986 Taken collectively, our data show the importance of the hydrophobic and charge transfer characteristics of hydrocarbon substrates in dictating the position of the cytochrome P-450 spin equilibrium, and as such, provides a rational molecular explanation based on sound chemical principles for the differential interaction of hydrocarbons with cytochrome P-450. Hydrocarbons 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 3742644-9 1986 Taken collectively, our data show the importance of the hydrophobic and charge transfer characteristics of hydrocarbon substrates in dictating the position of the cytochrome P-450 spin equilibrium, and as such, provides a rational molecular explanation based on sound chemical principles for the differential interaction of hydrocarbons with cytochrome P-450. Hydrocarbons 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 342-358 3742644-9 1986 Taken collectively, our data show the importance of the hydrophobic and charge transfer characteristics of hydrocarbon substrates in dictating the position of the cytochrome P-450 spin equilibrium, and as such, provides a rational molecular explanation based on sound chemical principles for the differential interaction of hydrocarbons with cytochrome P-450. Hydrocarbons 324-336 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 3742644-9 1986 Taken collectively, our data show the importance of the hydrophobic and charge transfer characteristics of hydrocarbon substrates in dictating the position of the cytochrome P-450 spin equilibrium, and as such, provides a rational molecular explanation based on sound chemical principles for the differential interaction of hydrocarbons with cytochrome P-450. Hydrocarbons 324-336 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 342-358 3018103-2 1986 The ability of different IFN species to induce xanthine oxidase correlated with their ability to depress liver cytochrome P-450-dependent drug metabolism, supporting the hypothesis that reactive oxygen metabolites generated by xanthine oxidase might be responsible for this impairment of liver function by IFN. Oxygen 195-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 3013312-0 1986 Direct formation of complexes between cytochrome P-450 and nitrosoarenes. nitrosoarenes 59-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 3013312-1 1986 The mechanism of the formation of the complexes between various nitrosobenzenes and cytochrome P-450 has been investigated. nitrosobenzene 64-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 3013312-3 1986 Nitrosobenzene (NOB) itself reacts with cytochrome P-450 in the iron(III) state, in the absence of any exogenous reducing agent, to produce the iron(II)-NOB complex. nitrosobenzene 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3013312-3 1986 Nitrosobenzene (NOB) itself reacts with cytochrome P-450 in the iron(III) state, in the absence of any exogenous reducing agent, to produce the iron(II)-NOB complex. nitrosobenzene 16-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3013312-3 1986 Nitrosobenzene (NOB) itself reacts with cytochrome P-450 in the iron(III) state, in the absence of any exogenous reducing agent, to produce the iron(II)-NOB complex. ferric sulfate 64-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3013312-3 1986 Nitrosobenzene (NOB) itself reacts with cytochrome P-450 in the iron(III) state, in the absence of any exogenous reducing agent, to produce the iron(II)-NOB complex. iron(ii)-nob 144-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 3013312-5 1986 The reduction of the iron may occur via ligand-induced oxidation of the axially bound thiolate of cytochrome P-450. Iron 21-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 3013312-5 1986 The reduction of the iron may occur via ligand-induced oxidation of the axially bound thiolate of cytochrome P-450. thiolate 86-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 3011017-0 1986 Oxidation of 1,4-dihydropyridines by prostaglandin synthase and the peroxidic function of cytochrome P-450. 1,4-dihydropyridine 13-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 3011017-2 1986 Oxidation of 1,4-dihydropyridines by the hydroperoxidic function of cytochrome P-450 and prostaglandin synthase was investigated using felodipine as a model substance. 1,4-dihydropyridine 13-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-111 3011017-2 1986 Oxidation of 1,4-dihydropyridines by the hydroperoxidic function of cytochrome P-450 and prostaglandin synthase was investigated using felodipine as a model substance. Felodipine 135-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-111 3960034-10 1986 These results of mutagenicity assay indicate that only the methyl derivatives of N-nitrosopropylamines, MHP and MOP are activated by the lung from 5 animal species and man, whereas the pancreas from all the tested animals did not activate the 7 N-nitrosopropylamines to mutagens, and that the phenobarbital-inducible major cytochrome P-450 in the lung of rodents is involved in the mutagenic activation of MHP. n-nitrosopropylamines 81-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 323-339 2874666-0 1986 The molecular mechanisms of two common polymorphisms of drug oxidation--evidence for functional changes in cytochrome P-450 isozymes catalysing bufuralol and mephenytoin oxidation. bufuralol 144-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-123 2874666-0 1986 The molecular mechanisms of two common polymorphisms of drug oxidation--evidence for functional changes in cytochrome P-450 isozymes catalysing bufuralol and mephenytoin oxidation. Mephenytoin 158-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-123 2874666-1 1986 Using the stereospecific metabolism of (+)- and (-)-bufuralol and (+)- and (-)-metoprolol as model reactions, we have characterized the enzymic deficiency of the debrisoquine/sparteine-type polymorphism by comparing kinetic data of subjects in vivo with their microsomal activities in vitro and with reconstituted activities of cytochrome P-450 isozymes purified from human liver. Debrisoquin 162-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 328-344 2874666-1 1986 Using the stereospecific metabolism of (+)- and (-)-bufuralol and (+)- and (-)-metoprolol as model reactions, we have characterized the enzymic deficiency of the debrisoquine/sparteine-type polymorphism by comparing kinetic data of subjects in vivo with their microsomal activities in vitro and with reconstituted activities of cytochrome P-450 isozymes purified from human liver. Sparteine 175-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 328-344 2874666-4 1986 A structural model of the active site of a cytochrome P-450 for stereospecific metabolism of bufuralol and other polymorphically metabolized substrates was constructed. bufuralol 93-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2874666-5 1986 Two cytochrome P-450 isozymes, P-450 buf I and P-450 buf II, both with MW 50,000 Da, were purified from human liver on the basis of their ability to metabolize bufuralol to 1"-hydroxy-bufuralol. bufuralol 160-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2874666-5 1986 Two cytochrome P-450 isozymes, P-450 buf I and P-450 buf II, both with MW 50,000 Da, were purified from human liver on the basis of their ability to metabolize bufuralol to 1"-hydroxy-bufuralol. Ro 03-7410 173-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2874666-13 1986 A cytochrome P-450 with high activity for mephenytoin 4-hydroxylation was purified from human liver. Mephenytoin 42-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-18 3739365-5 1986 By monitoring the hydroxylation of DMI in various fractions during separation and purification of cytochrome P-450 from human liver microsomes we have purified a cytochrome P-450 which efficiently hydroxylates this drug. Desipramine 35-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 3739365-5 1986 By monitoring the hydroxylation of DMI in various fractions during separation and purification of cytochrome P-450 from human liver microsomes we have purified a cytochrome P-450 which efficiently hydroxylates this drug. Desipramine 35-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-178 3739365-8 1986 This is probably the major debrisoquine hydroxylating cytochrome P-450 in man. Debrisoquin 27-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 3010988-0 1986 DNA strand scission by hemin-thiolate complexes as models of cytochrome P-450. hemin-thiolate 23-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 3010988-1 1986 Hemin-thiolate complexes, as chemical models for cytochrome P-450 monooxygenases, have been shown to cause strand scission of DNA. hemin-thiolate 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 3697359-2 1986 Cytochrome P-450 I-c and cytochrome P-450 II-d, both obtained from the hepatic microsome of polychlorinated biphenyl-treated rats, were used as P-450 type and P-448 type, respectively, and benzo[a]pyrene and 7-ethoxycoumarin were used as substrate. Polychlorinated Biphenyls 92-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3697359-2 1986 Cytochrome P-450 I-c and cytochrome P-450 II-d, both obtained from the hepatic microsome of polychlorinated biphenyl-treated rats, were used as P-450 type and P-448 type, respectively, and benzo[a]pyrene and 7-ethoxycoumarin were used as substrate. Polychlorinated Biphenyls 92-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 3697359-5 1986 Differences in the conformational change were observed between the two enzymes, and the conformational change of cytochrome P-450 II-d using benzo[a]pyrene as a substrate was different from that using 7-ethoxycoumarin. Benzo(a)pyrene 141-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 3697359-5 1986 Differences in the conformational change were observed between the two enzymes, and the conformational change of cytochrome P-450 II-d using benzo[a]pyrene as a substrate was different from that using 7-ethoxycoumarin. 7-ethoxycoumarin 201-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 3697359-6 1986 The results suggest that the substrate-binding sites of the two enzymes differ in position toward the heme, and that there are at least two different substrate sites in cytochrome P-450 II-d, one for benzo[a]pyrene and another for 7-ethoxycoumarin. Heme 102-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 3697359-6 1986 The results suggest that the substrate-binding sites of the two enzymes differ in position toward the heme, and that there are at least two different substrate sites in cytochrome P-450 II-d, one for benzo[a]pyrene and another for 7-ethoxycoumarin. Benzo(a)pyrene 200-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 3697359-6 1986 The results suggest that the substrate-binding sites of the two enzymes differ in position toward the heme, and that there are at least two different substrate sites in cytochrome P-450 II-d, one for benzo[a]pyrene and another for 7-ethoxycoumarin. 7-ethoxycoumarin 231-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 3514607-0 1986 Characterization of rat and human liver microsomal cytochrome P-450 forms involved in nifedipine oxidation, a prototype for genetic polymorphism in oxidative drug metabolism. Nifedipine 86-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 3514607-5 1986 Nifedipine oxidation has been shown to be catalyzed by cytochrome P-450 (P-450) enzymes. Nifedipine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 3964299-1 1986 Clofibrate, an antilipidemic drug that acts by a still obscure mechanism, is known to specifically increase up to 30-fold the activity of the hepatic cytochrome P-450 isozyme that omega-hydroxlates lauric acid. Clofibrate 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 3964299-1 1986 Clofibrate, an antilipidemic drug that acts by a still obscure mechanism, is known to specifically increase up to 30-fold the activity of the hepatic cytochrome P-450 isozyme that omega-hydroxlates lauric acid. lauric acid 198-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 3718550-0 1986 [Hydroxylation of steroids in a dienzyme system consisting of cytochrome P-450 and immobilized adrenodoxin]. Steroids 18-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3754494-0 1986 Metabolic nitrite formation from N-nitrosamines: are there other pathways than reductive denitrosation by cytochrome P-450? Nitrites 10-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 3711591-1 1986 Effect of acute doses of technical grade of dichloro diphenyl trichloro ethane (DDT) and piperonyl butoxide (PB) on hepatic microsomal cytochrome P450 (Cyt. DDT 44-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 3711591-1 1986 Effect of acute doses of technical grade of dichloro diphenyl trichloro ethane (DDT) and piperonyl butoxide (PB) on hepatic microsomal cytochrome P450 (Cyt. DDT 80-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 3711591-1 1986 Effect of acute doses of technical grade of dichloro diphenyl trichloro ethane (DDT) and piperonyl butoxide (PB) on hepatic microsomal cytochrome P450 (Cyt. Piperonyl Butoxide 89-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 3711591-1 1986 Effect of acute doses of technical grade of dichloro diphenyl trichloro ethane (DDT) and piperonyl butoxide (PB) on hepatic microsomal cytochrome P450 (Cyt. Piperonyl Butoxide 109-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 3517618-0 1986 Identification of a human liver cytochrome P-450 homologous to the major isosafrole-inducible cytochrome P-450 in the rat. isosafrole 73-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 3517618-0 1986 Identification of a human liver cytochrome P-450 homologous to the major isosafrole-inducible cytochrome P-450 in the rat. isosafrole 73-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 3517618-6 1986 We conclude that, in man, there is a cytochrome P-450 family composed of two isozymes (HLc and HLd) that are immunochemically and structurally related to the 3-methylcholanthrene-inducible family observed in several other species. Methylcholanthrene 158-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 3964273-2 1986 In addition, a cysteine containing peptide with a sequence homologous to those of peptides containing the cysteine residue which was suggested to provide the proximal thiolate ligand to the heme in other cytochrome p-450 isozymes, was identified. Cysteine 15-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-220 3964273-2 1986 In addition, a cysteine containing peptide with a sequence homologous to those of peptides containing the cysteine residue which was suggested to provide the proximal thiolate ligand to the heme in other cytochrome p-450 isozymes, was identified. Peptides 35-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-220 3964273-2 1986 In addition, a cysteine containing peptide with a sequence homologous to those of peptides containing the cysteine residue which was suggested to provide the proximal thiolate ligand to the heme in other cytochrome p-450 isozymes, was identified. Peptides 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-220 3964273-2 1986 In addition, a cysteine containing peptide with a sequence homologous to those of peptides containing the cysteine residue which was suggested to provide the proximal thiolate ligand to the heme in other cytochrome p-450 isozymes, was identified. Cysteine 106-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-220 3964273-2 1986 In addition, a cysteine containing peptide with a sequence homologous to those of peptides containing the cysteine residue which was suggested to provide the proximal thiolate ligand to the heme in other cytochrome p-450 isozymes, was identified. thiolate 167-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-220 3964273-2 1986 In addition, a cysteine containing peptide with a sequence homologous to those of peptides containing the cysteine residue which was suggested to provide the proximal thiolate ligand to the heme in other cytochrome p-450 isozymes, was identified. Heme 190-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-220 3511825-4 1986 The possible causes for the injury from ostensibly nontoxic drug levels appear to be either the induction by chronic alcohol intake of the cytochrome P-450 system responsible for converting acetaminophen to a toxic metabolite, or the effect of alcoholism and the associated malnutrition in reducing the glutathione concentration, responsible normally for preventing hepatotoxicity by conjugation with the toxic metabolite. Alcohols 117-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 3511825-4 1986 The possible causes for the injury from ostensibly nontoxic drug levels appear to be either the induction by chronic alcohol intake of the cytochrome P-450 system responsible for converting acetaminophen to a toxic metabolite, or the effect of alcoholism and the associated malnutrition in reducing the glutathione concentration, responsible normally for preventing hepatotoxicity by conjugation with the toxic metabolite. Acetaminophen 190-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 3511825-4 1986 The possible causes for the injury from ostensibly nontoxic drug levels appear to be either the induction by chronic alcohol intake of the cytochrome P-450 system responsible for converting acetaminophen to a toxic metabolite, or the effect of alcoholism and the associated malnutrition in reducing the glutathione concentration, responsible normally for preventing hepatotoxicity by conjugation with the toxic metabolite. Glutathione 303-314 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 3485990-0 1986 Inhibition of human liver cytochrome P-450 by omeprazole. Omeprazole 46-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 3485990-1 1986 The effects of omeprazole on cytochrome P-450 mediated 7-ethoxycoumarin deethylation were studied in human liver microsomes. Omeprazole 15-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 3485990-1 1986 The effects of omeprazole on cytochrome P-450 mediated 7-ethoxycoumarin deethylation were studied in human liver microsomes. 7-ethoxycoumarin 55-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 3485990-3 1986 The results are further evidence that the previously reported prolongation of the half-life of diazepam by omeprazole in vivo is due to inhibition of cytochrome P-450 monooxygenases. Diazepam 95-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 3485990-3 1986 The results are further evidence that the previously reported prolongation of the half-life of diazepam by omeprazole in vivo is due to inhibition of cytochrome P-450 monooxygenases. Omeprazole 107-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 3085302-0 1986 [Cytochrome P-450-dependent pathway of oxidation of arachidonic acid and its metabolites]. Arachidonic Acid 52-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 1-17 3085302-1 1986 The experimental data on the role of cytochrome P-450 in the metabolism of arachidonic acid and protanoids in different organs and tissues are analyzed. Arachidonic Acid 75-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 3085302-1 1986 The experimental data on the role of cytochrome P-450 in the metabolism of arachidonic acid and protanoids in different organs and tissues are analyzed. protanoids 96-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 3085302-3 1986 A conclusion is drawn that cytochrome P-450 takes an active part not only in oxidation of different xenobiotics and steroid hormones but also in the synthesis and decomposition of prostaglandins--the most important cell regulators. Steroids 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 3085302-3 1986 A conclusion is drawn that cytochrome P-450 takes an active part not only in oxidation of different xenobiotics and steroid hormones but also in the synthesis and decomposition of prostaglandins--the most important cell regulators. Prostaglandins 180-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 3004505-12 1986 The PAH produced a 2-fold increase in spectroscopically detectable cytochrome P-450 levels in NCI-H322. Polycyclic Aromatic Hydrocarbons 4-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3004505-12 1986 The PAH produced a 2-fold increase in spectroscopically detectable cytochrome P-450 levels in NCI-H322. 4-Bromo-3-fluoroaniline 98-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3947400-0 1986 Induction of cytochrome P-450 by alcohols and 4-substituted pyrazoles. Alcohols 33-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 3947400-0 1986 Induction of cytochrome P-450 by alcohols and 4-substituted pyrazoles. 4-substituted pyrazoles 46-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 3947400-2 1986 A comparison was made between 4-substituted pyrazoles and short-chain alcohols as inducers of cytochrome P-450. 4-substituted pyrazoles 30-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 3947400-2 1986 A comparison was made between 4-substituted pyrazoles and short-chain alcohols as inducers of cytochrome P-450. Alcohols 70-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 3947400-3 1986 A quantitative structure-activity analysis of the data led to the following equations: (I) Pyrazoles: Log 1/C = 0.85 (+/- 0.21) Log P + 1.93 (+/- 0.38), r = 0.970 (II) Alcohols: Log 1/C = 0.78 (+/- 0.14) Log P + 1.46 (+/- 0.13), r = 0.988 where C is the concentration that caused a 50% increase in cytochrome P-450, is the partition coefficient between octanol and water, and r is the correlation coefficient. Pyrazoles 91-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-314 3510912-4 1986 The liver contains the most important of these detoxification systems: the cytochrome P-450-dependent mixed function oxidase (MFO) and several conjugation enzymes, e.g., sulfotransferase, glucuronyl transferase, and glutathione transferase, which convert lipophilic compounds to more water-soluble products to enhance their excretion. Water 284-289 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 3510912-8 1986 Diets restricted in calories, protein, or essential fatty acids, as well as those having low quality protein or high sugar content, can affect the component enzymes, cytochrome P-450 and the cytochrome P-450 reductase, and the MFO activity toward a variety of drugs. Fatty Acids, Essential 42-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-182 3510912-8 1986 Diets restricted in calories, protein, or essential fatty acids, as well as those having low quality protein or high sugar content, can affect the component enzymes, cytochrome P-450 and the cytochrome P-450 reductase, and the MFO activity toward a variety of drugs. Fatty Acids, Essential 42-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-207 3510912-8 1986 Diets restricted in calories, protein, or essential fatty acids, as well as those having low quality protein or high sugar content, can affect the component enzymes, cytochrome P-450 and the cytochrome P-450 reductase, and the MFO activity toward a variety of drugs. Sugars 117-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-182 3510912-8 1986 Diets restricted in calories, protein, or essential fatty acids, as well as those having low quality protein or high sugar content, can affect the component enzymes, cytochrome P-450 and the cytochrome P-450 reductase, and the MFO activity toward a variety of drugs. Sugars 117-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-207 3511095-2 1986 Such studies have revealed the important role that the central metal atom plays in determining the physiological and pharmacological properties of metal-porphyrin complexes; and they have demonstrated that the form in which animals and humans are exposed to trace metals, i.e., inorganic, organified, porphyrin-chelated, etc., can be of great importance in determining the biological responses that such elements elicit, especially with respect to actions on heme synthesis and degradation and cytochrome P-450 formation and function. Metals 147-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 494-510 3511095-2 1986 Such studies have revealed the important role that the central metal atom plays in determining the physiological and pharmacological properties of metal-porphyrin complexes; and they have demonstrated that the form in which animals and humans are exposed to trace metals, i.e., inorganic, organified, porphyrin-chelated, etc., can be of great importance in determining the biological responses that such elements elicit, especially with respect to actions on heme synthesis and degradation and cytochrome P-450 formation and function. Porphyrins 153-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 494-510 3082822-0 1986 A comparative study of the mutagenic activation of N-nitrosopropylamines by various animal species and man: evidence for a cytochrome P-450 dependent reaction. n-nitrosopropylamines 51-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 3082822-8 1986 These results demonstrated that the phenobarbital-inducible major cytochrome P-450 in animal and human livers is involved in the mutagenic activation of the N-nitrosopropylamines. Phenobarbital 36-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 3082822-8 1986 These results demonstrated that the phenobarbital-inducible major cytochrome P-450 in animal and human livers is involved in the mutagenic activation of the N-nitrosopropylamines. n-nitrosopropylamines 157-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 3961272-2 1986 PEG-6000 and ammonium sulfate fractionation of human granulocyte post-mitochondrial supernatant (S1) fraction resulted in 3.9 and 2.25 fold purification of cytochrome P-450 respectively. Polyethylene Glycol 6000 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 3961272-2 1986 PEG-6000 and ammonium sulfate fractionation of human granulocyte post-mitochondrial supernatant (S1) fraction resulted in 3.9 and 2.25 fold purification of cytochrome P-450 respectively. Ammonium Sulfate 13-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 3961272-3 1986 On the other hand, granulocyte S1 fractions subjected to noctylamino sepharose 4B and DEAE cellulose column chromatography separately revealed about 11 fold purification of cytochrome P-450. noctylamino sepharose 4b 57-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-189 3961272-3 1986 On the other hand, granulocyte S1 fractions subjected to noctylamino sepharose 4B and DEAE cellulose column chromatography separately revealed about 11 fold purification of cytochrome P-450. DEAE-Cellulose 86-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-189 3080992-1 1986 Water-soluble carbodiimide-catalyzed cross-linking of purified cytochrome P-450 LM2, cytochrome b5, and NADPH-cytochrome P-450 reductase was used to identify stable complexes formed between these proteins. Water 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 3080992-1 1986 Water-soluble carbodiimide-catalyzed cross-linking of purified cytochrome P-450 LM2, cytochrome b5, and NADPH-cytochrome P-450 reductase was used to identify stable complexes formed between these proteins. Carbodiimides 14-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 3955002-0 1986 Ligand and halide binding properties of chloroperoxidase: peroxidase-type active site heme environment with cytochrome P-450 type endogenous axial ligand and spectroscopic properties. halide 11-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 3955002-0 1986 Ligand and halide binding properties of chloroperoxidase: peroxidase-type active site heme environment with cytochrome P-450 type endogenous axial ligand and spectroscopic properties. Heme 86-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 3955002-4 1986 All of the ligands except for the halides, nitrate, and acetate form exclusively low-spin complexes in analogy to results obtained with the spectroscopically related protein, cytochrome P-450-CAM [Sono, M., & Dawson, J.H. Nitrates 43-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-191 3955002-4 1986 All of the ligands except for the halides, nitrate, and acetate form exclusively low-spin complexes in analogy to results obtained with the spectroscopically related protein, cytochrome P-450-CAM [Sono, M., & Dawson, J.H. Acetates 56-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-191 3532707-4 1986 In our laboratory we have demonstrated that DBE is metabolized by both cytochrome P-450 and GSH S-transferases. Ethylene Dibromide 44-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 3532707-5 1986 Although the cytochrome P-450 metabolite is reactive and will covalently bind to protein and nucleic acid to some extent, and the GSH S-transferase system conjugates it and under conditions of low DBE exposure is able to detoxify it. Ethylene Dibromide 197-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 3591537-7 1986 Zinc deficiency is known to increase the cytochrome P-450-dependent metabolism of methylbenzylnitrosamine (MBN), an esophageal carcinogen of this class. nitrosobenzylmethylamine 82-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 3591537-7 1986 Zinc deficiency is known to increase the cytochrome P-450-dependent metabolism of methylbenzylnitrosamine (MBN), an esophageal carcinogen of this class. nitrosobenzylmethylamine 107-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 3954830-3 1986 The data suggest that most of the ethanol metabolism by blood-monocyte-derived macrophages is mediated via the cytochrome-P-450-dependent microsomal ethanol-oxidising system. Ethanol 34-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 3954830-3 1986 The data suggest that most of the ethanol metabolism by blood-monocyte-derived macrophages is mediated via the cytochrome-P-450-dependent microsomal ethanol-oxidising system. Ethanol 149-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 3790107-0 1986 The molecular biology of the polycyclic aromatic hydrocarbon inducible cytochrome P-450; the past is prologue. Polycyclic Aromatic Hydrocarbons 29-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 3940522-0 1986 Benznidazole: nitroreduction and inhibition of cytochrome P-450 in chemosensitization of tumour response to cytotoxic drugs. benzonidazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 3955103-6 1986 The experimental results suggest that the activity and structure of cytochrome P-450 conformers is affected by the lipid microenvironment, type I substrates and Triton N-101. Triton N 101 161-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 3094971-6 1986 Aminoglutethimide, however, blocks other cytochrome P-450-mediated steroid hydroxylations, requires concomitant glucocorticoid administration, and is associated with initial side effects. Aminoglutethimide 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 3094971-6 1986 Aminoglutethimide, however, blocks other cytochrome P-450-mediated steroid hydroxylations, requires concomitant glucocorticoid administration, and is associated with initial side effects. Steroids 67-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 3539534-10 1986 When drugs such as phenobarbital are administered to animals, there is a rapid induction in the liver of both cytochrome P-450 and ALV-synthase. Phenobarbital 19-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-143 3539534-11 1986 It is proposed that the heme pool governing ALV-synthase levels is lowered by the increased heme demand due to cytochrome P-450 apoprotein formation. Heme 24-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 3539534-11 1986 It is proposed that the heme pool governing ALV-synthase levels is lowered by the increased heme demand due to cytochrome P-450 apoprotein formation. Heme 92-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 3539534-18 1986 Attacks of the disease are commonly precipitated by drugs which induce cytochrome P-450, and the uncontrolled accumulation of ALV-synthase which accompanies these attacks results from the combined action of the block in the heme pathway and the increased cytochrome P-450 levels. Heme 224-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 255-271 2868861-2 1986 In agreement with earlier studies, cobalt chloride treatment produced a 1.5-fold increase in hepatic glutathione levels and a decrease in the cytochrome P-450-dependent oxidative metabolism of acetaminophen. cobaltous chloride 35-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-158 2868861-2 1986 In agreement with earlier studies, cobalt chloride treatment produced a 1.5-fold increase in hepatic glutathione levels and a decrease in the cytochrome P-450-dependent oxidative metabolism of acetaminophen. Acetaminophen 193-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-158 2868861-5 1986 Collectively, the data suggest that the protective effect of cobalt chloride treatment on acetaminophen hepatotoxicity results from a combination of the suppression of the toxic pathway of cytochrome P-450 oxidation, an increase in the protective capacity of glutathione, and an enhancement of the nontoxic pathway of glucuronidation. cobaltous chloride 61-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-205 2868861-5 1986 Collectively, the data suggest that the protective effect of cobalt chloride treatment on acetaminophen hepatotoxicity results from a combination of the suppression of the toxic pathway of cytochrome P-450 oxidation, an increase in the protective capacity of glutathione, and an enhancement of the nontoxic pathway of glucuronidation. Acetaminophen 90-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-205 3948674-0 1986 [The role of cytochrome P-450 in the activation of phosphatidyl choline hydrolysis by phospholipase A2]. Phosphatidylcholines 51-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 3549274-2 1986 The pathway of biosynthesis of these steroids from cholesterol involves a sequence of transformations using cytochrome P-450 enzymes. Steroids 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 3549274-2 1986 The pathway of biosynthesis of these steroids from cholesterol involves a sequence of transformations using cytochrome P-450 enzymes. Cholesterol 51-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 3549274-3 1986 The hypothesis presented here is that damage to cytochrome P-450 enzymes on interaction with certain steroids, synthesized by the adrenal cortex itself, may be of pathological and perhaps physiological importance. Steroids 101-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 3549274-4 1986 The interaction between cytochrome P-450 enzymes and these steroids, which act as pseudosubstrates, may form part of the pathogenesis of some steroidogenic enzyme deficiencies, with consequent overproduction of precursor steroids, leading to mineralocorticoid or androgen excess. Steroids 59-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 3549274-4 1986 The interaction between cytochrome P-450 enzymes and these steroids, which act as pseudosubstrates, may form part of the pathogenesis of some steroidogenic enzyme deficiencies, with consequent overproduction of precursor steroids, leading to mineralocorticoid or androgen excess. Steroids 221-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 3549274-7 1986 The high content of antioxidant compounds in the adrenal cortex, principally ascorbate, may serve to protect cytochrome P-450 enzymes from the damaging effects of oxygen radical species formed as a result of cytochrome P-450/pseudosubstrate interactions. Ascorbic Acid 77-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 3549274-7 1986 The high content of antioxidant compounds in the adrenal cortex, principally ascorbate, may serve to protect cytochrome P-450 enzymes from the damaging effects of oxygen radical species formed as a result of cytochrome P-450/pseudosubstrate interactions. Ascorbic Acid 77-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 208-224 3549274-7 1986 The high content of antioxidant compounds in the adrenal cortex, principally ascorbate, may serve to protect cytochrome P-450 enzymes from the damaging effects of oxygen radical species formed as a result of cytochrome P-450/pseudosubstrate interactions. Oxygen 163-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 3549274-7 1986 The high content of antioxidant compounds in the adrenal cortex, principally ascorbate, may serve to protect cytochrome P-450 enzymes from the damaging effects of oxygen radical species formed as a result of cytochrome P-450/pseudosubstrate interactions. Oxygen 163-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 208-224 3709653-3 1986 2-hydroxylation of DMI is probably mediated by debrisoquine hydroxylase, a cytochrome P-450 isozyme that is monogenically controlled. Desipramine 19-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 3562262-1 1986 Thiolate-hemin complexes as chemical models for cytochrome P-450 have been shown to cause cleavage of DNA. thiolate-hemin 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 3739442-0 1986 Effect of phenobarbital on the contents of microsomal cytochrome P-450 and levels of glutathione (GSH) in the liver of various animal species. Phenobarbital 10-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 2869518-0 1985 Cytochrome P450: substrate and prosthetic-group free radicals generated during the enzymatic cycle. Free Radicals 48-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 4083898-1 1985 Aromatase cytochrome P-450 (P-450AROM) was partially purified from human placental microsomes by hydrophobic affinity chromatography using Phenyl-Sepharose and ion-exchange chromatography on DEAE-cellulose. phenyl-sepharose 139-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 4083898-1 1985 Aromatase cytochrome P-450 (P-450AROM) was partially purified from human placental microsomes by hydrophobic affinity chromatography using Phenyl-Sepharose and ion-exchange chromatography on DEAE-cellulose. DEAE-Cellulose 191-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 4083898-3 1985 When the cytochrome P-450-enriched fractions were subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis and stained with Coomassie blue, there was an enrichment of two proteins having apparent molecular weights of 50,000 and 55,000. Sodium Dodecyl Sulfate 63-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 4083898-3 1985 When the cytochrome P-450-enriched fractions were subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis and stained with Coomassie blue, there was an enrichment of two proteins having apparent molecular weights of 50,000 and 55,000. polyacrylamide gels 86-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 4083898-3 1985 When the cytochrome P-450-enriched fractions were subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis and stained with Coomassie blue, there was an enrichment of two proteins having apparent molecular weights of 50,000 and 55,000. Coomassie blue 138-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 4083898-10 1985 The finding that cytochrome P-450 and the 55-kDa protein were selectively retained by the affinity column and eluted with NaCl (2 M) and glycine (0.2 M, pH 3.0) and that this fraction contained aromatase activity upon reconstitution with purified NADPH-cytochrome P-450 reductase and phospholipid, is indicative that the 55-kDa protein is indeed cytochrome P-450AROM. Sodium Chloride 122-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 4083898-10 1985 The finding that cytochrome P-450 and the 55-kDa protein were selectively retained by the affinity column and eluted with NaCl (2 M) and glycine (0.2 M, pH 3.0) and that this fraction contained aromatase activity upon reconstitution with purified NADPH-cytochrome P-450 reductase and phospholipid, is indicative that the 55-kDa protein is indeed cytochrome P-450AROM. Glycine 137-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 3007100-0 1985 Model studies in cytochrome P-450-mediated toxicity of halogenated compounds: radical processes involving iron porphyrins. iron porphyrins 106-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 3007100-1 1985 Haloalkane toxicity originates from attack on biological targets by reactive intermediates derived from haloalkane metabolism by a hemoprotein, cytochrome P-450. haloalkane 104-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 3007100-3 1985 The reactions of iron porphyrin--a model for cytochrome P-450--with various carbon-centered and peroxyl radicals generated by pulse radiolysis are examined. iron porphyrin 17-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 3007100-3 1985 The reactions of iron porphyrin--a model for cytochrome P-450--with various carbon-centered and peroxyl radicals generated by pulse radiolysis are examined. Carbon 76-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 3007100-3 1985 The reactions of iron porphyrin--a model for cytochrome P-450--with various carbon-centered and peroxyl radicals generated by pulse radiolysis are examined. perhydroxyl radical 96-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 3007100-6 1985 The importance of local oxygen concentration and structural arrangement of fatty acids around cytochrome P-450 is emphasized. Fatty Acids 75-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 4061517-1 1985 Variations in cytochrome P-450 levels may influence the responsiveness of uterine and breast tissue as well as carcinomas to endocrine therapy and may be of particular importance with agents such as tamoxifen (Nolvadex) where hydroxylation is known to alter therapeutic activities. Tamoxifen 199-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 4061517-1 1985 Variations in cytochrome P-450 levels may influence the responsiveness of uterine and breast tissue as well as carcinomas to endocrine therapy and may be of particular importance with agents such as tamoxifen (Nolvadex) where hydroxylation is known to alter therapeutic activities. Tamoxifen 210-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 4061517-3 1985 Cyclohexane served as a substrate for several forms of cytochrome P-450. Cyclohexane 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). Sodium Azide 23-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). N,N-dimethylaniline 166-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). N,N-dimethylaniline 183-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). Aminopyrine 189-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). Benzphetamine 207-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). Benzphetamine 222-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). 4-nitroanisole 228-242 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). 4-nitroanisole 244-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). aniline 272-281 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-1 1985 It has been shown that sodium azide, a catalase inhibitor, accelerates the inactivation of cytochrome P-450 in reactions of hydroxylation of type I substrates, e.g., dimethylaniline (DMA), aminopyrine (AP), benzphetamine (BPh), p-nitroanisole (p-Na) and type II substrates--aniline (AN). an 283-285 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 4063403-4 1985 Studies on the mechanism of cytochrome P-450 inactivation demonstrated that hydrogen peroxide formed via the NADPH-dependent hydroxylase cycle exhibits an inactivating effect towards the enzyme. Hydrogen Peroxide 76-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 4063403-5 1985 The value of inactivation constant for cytochrome P-450 in hydroxylation reactions proceeding in the presence of sodium azide coincides with that of enzyme inactivation in the NADPH-oxidase and glucose oxidase systems generating H2O2. Sodium Azide 113-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 4063403-5 1985 The value of inactivation constant for cytochrome P-450 in hydroxylation reactions proceeding in the presence of sodium azide coincides with that of enzyme inactivation in the NADPH-oxidase and glucose oxidase systems generating H2O2. Hydrogen Peroxide 229-233 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 4053486-5 1985 These results support our hypothesis that the mephenytoin polymorphism is caused by a partial or complete absence or inactivity of a cytochrome P-450 isozyme with high affinity for S-mephenytoin. Mephenytoin 46-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-149 4053486-5 1985 These results support our hypothesis that the mephenytoin polymorphism is caused by a partial or complete absence or inactivity of a cytochrome P-450 isozyme with high affinity for S-mephenytoin. Mephenytoin 181-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-149 4064917-10 1985 These results implicate cimetidine binding to the cytochrome P-450 system as the sole mechanism responsible for inhibition of the systemic clearance of co-administered drugs metabolized by the liver. Cimetidine 24-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 3912769-0 1985 Cytochrome P-450 dependent pathways in corticosteroid hormones biosynthesis. corticosteroid hormones 39-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 4095131-0 1985 [Elimination of caffeine and metamizol in in vivo characterization of cytochrome P-450 dependent biotransformation reactions in aged humans]. Caffeine 16-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 4095131-0 1985 [Elimination of caffeine and metamizol in in vivo characterization of cytochrome P-450 dependent biotransformation reactions in aged humans]. Dipyrone 29-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 4095131-2 1985 From the elimination velocity of these model substances conclusions concerning the activity of 3-methylcholanthrene inducible (caffeine elimination) and of phenobarbital inducible (metamizol elimination) isoenzymes of cytochrome P-450 are drawn. Methylcholanthrene 95-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 218-234 4095131-2 1985 From the elimination velocity of these model substances conclusions concerning the activity of 3-methylcholanthrene inducible (caffeine elimination) and of phenobarbital inducible (metamizol elimination) isoenzymes of cytochrome P-450 are drawn. Phenobarbital 156-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 218-234 4095131-2 1985 From the elimination velocity of these model substances conclusions concerning the activity of 3-methylcholanthrene inducible (caffeine elimination) and of phenobarbital inducible (metamizol elimination) isoenzymes of cytochrome P-450 are drawn. Dipyrone 181-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 218-234 4095132-1 1985 Caffeine is mainly metabolized by 3-methylcholanthrene-inducible cytochrome P-450 (P-450MC) and metamizol (noramidopyrine methanesulfonate sodium) is mainly metabolized by phenobarbital-inducible cytochrome P-450 (P-450PB). Caffeine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 4095132-1 1985 Caffeine is mainly metabolized by 3-methylcholanthrene-inducible cytochrome P-450 (P-450MC) and metamizol (noramidopyrine methanesulfonate sodium) is mainly metabolized by phenobarbital-inducible cytochrome P-450 (P-450PB). Methylcholanthrene 34-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 4095132-2 1985 That"s why the half life of caffeine and the elimination of the main metabolites of metamizol were used as parameters in vivo characterizing both groups of the cytochrome P-450-complex. Caffeine 28-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 4095132-2 1985 That"s why the half life of caffeine and the elimination of the main metabolites of metamizol were used as parameters in vivo characterizing both groups of the cytochrome P-450-complex. Dipyrone 84-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 2997155-3 1985 To develop pharmacologically useful inhibitors of the LTB4 omega-hydroxylase in human PMN, we devised a general scheme for synthesizing terminal acetylenic fatty acids based on the "acetylenic zipper" reaction. acetylenic fatty acids 145-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-76 2997155-4 1985 We found that the LTB4 omega-hydroxylase in intact PMN and in PMN sonicates is inactivated in a concentration-dependent fashion by terminal acetylenic analogues of lauric, palmitic, and stearic acids (i.e. 11-dodecynoic, 15-hexadecynoic, and 17-octadecynoic acids). acetylenic 140-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-40 2997155-4 1985 We found that the LTB4 omega-hydroxylase in intact PMN and in PMN sonicates is inactivated in a concentration-dependent fashion by terminal acetylenic analogues of lauric, palmitic, and stearic acids (i.e. 11-dodecynoic, 15-hexadecynoic, and 17-octadecynoic acids). Stearic Acids 186-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-40 2997155-4 1985 We found that the LTB4 omega-hydroxylase in intact PMN and in PMN sonicates is inactivated in a concentration-dependent fashion by terminal acetylenic analogues of lauric, palmitic, and stearic acids (i.e. 11-dodecynoic, 15-hexadecynoic, and 17-octadecynoic acids). -dodecynoic 208-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-40 2997155-4 1985 We found that the LTB4 omega-hydroxylase in intact PMN and in PMN sonicates is inactivated in a concentration-dependent fashion by terminal acetylenic analogues of lauric, palmitic, and stearic acids (i.e. 11-dodecynoic, 15-hexadecynoic, and 17-octadecynoic acids). 17-octadecynoic acid 242-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-40 2997155-5 1985 Consistent with a suicidal process, inactivation of the LTB4 omega-hydroxylase requires molecular oxygen and NADPH, is time-dependent, and follows pseudo-first-order kinetics. Oxygen 98-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-78 2997155-5 1985 Consistent with a suicidal process, inactivation of the LTB4 omega-hydroxylase requires molecular oxygen and NADPH, is time-dependent, and follows pseudo-first-order kinetics. NADP 109-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-78 3000277-1 1985 We have recently isolated a cloned cDNA coding for a cytochrome P-450 of human liver microsomal membranes, which corresponds to a major phenobarbital-inducible cytochrome P-450 of rat liver. Phenobarbital 136-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 3000277-1 1985 We have recently isolated a cloned cDNA coding for a cytochrome P-450 of human liver microsomal membranes, which corresponds to a major phenobarbital-inducible cytochrome P-450 of rat liver. Phenobarbital 136-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 4052098-3 1985 This epitope was also present on cytochrome P-450 PCN-E (pregnenolone-16 alpha-carbonitrile induced) from rat liver microsomes, but this isozyme differed from the human P-450(5) by its molecular weight. pcn-e 50-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 4052098-3 1985 This epitope was also present on cytochrome P-450 PCN-E (pregnenolone-16 alpha-carbonitrile induced) from rat liver microsomes, but this isozyme differed from the human P-450(5) by its molecular weight. Pregnenolone Carbonitrile 57-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 4074778-0 1985 [A shunted system of electron transport from NAD(P)H to cholesterol-hydroxylating cytochrome P-450 in adrenal cortex mitochondria]. nad(p)h 45-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 4074778-0 1985 [A shunted system of electron transport from NAD(P)H to cholesterol-hydroxylating cytochrome P-450 in adrenal cortex mitochondria]. Cholesterol 56-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 4074778-7 1985 Phenazine methosulfate can reduce microsomal cytochrome P-450 LM2 and b5, but not cytochrome P-450 LM4. Methylphenazonium Methosulfate 0-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 3000715-0 1985 Cloning and isolation of human cytochrome P-450 cDNAs homologous to dioxin-inducible rabbit mRNAs encoding P-450 4 and P-450 6. Dioxins 68-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 4052083-5 1985 Activities of 7-ethoxycoumarin O-deethylase and benzo(a)pyrene hydroxylase in livers but not in adrenals were inhibited by the anti-P-450 HFLa antibodies, probably suggesting that distinct forms of cytochrome P-450 are responsible for the oxidations in livers and adrenals. 7-ethoxycoumarin 14-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-214 3839750-0 1985 Identification and quantification of a messenger ribonucleic acid induced by polynuclear aromatic hydrocarbons--using a cloned human cytochrome P-450 gene. Polycyclic Aromatic Hydrocarbons 77-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-149 3839750-1 1985 We have isolated four overlapping human genomic clones associated with the polynuclear aromatic hydrocarbon-induced form of cytochrome P-450. Hydrocarbons, Aromatic 87-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 3935158-0 1985 Kinetics of cytochrome P-450 reduction: evidence for faster reduction of the high-spin ferric state. Ferric enterobactin ion 87-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 3935158-10 1985 A mathematical expression of the fractional content of high-spin cytochrome P-450 was obtained by comparing the ratio of the initial rate of change in the fraction of total oxidized cytochrome remaining to the initial rate of change in the fraction of high-spin ferric P-450 remaining. ferric p 262-270 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 2865110-9 1985 When compared with previous data relating to theophylline, the results suggest that theobromine 3-demethylation is mediated by the same form(s) of cytochrome P-450 involved in theophylline demethylation, while a second form(s) of cytochrome P-450 is involved in theobromine 7-demethylation and theophylline 8-hydroxylation. Theophylline 45-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2865110-9 1985 When compared with previous data relating to theophylline, the results suggest that theobromine 3-demethylation is mediated by the same form(s) of cytochrome P-450 involved in theophylline demethylation, while a second form(s) of cytochrome P-450 is involved in theobromine 7-demethylation and theophylline 8-hydroxylation. Theobromine 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2865110-9 1985 When compared with previous data relating to theophylline, the results suggest that theobromine 3-demethylation is mediated by the same form(s) of cytochrome P-450 involved in theophylline demethylation, while a second form(s) of cytochrome P-450 is involved in theobromine 7-demethylation and theophylline 8-hydroxylation. Theobromine 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-246 2865110-9 1985 When compared with previous data relating to theophylline, the results suggest that theobromine 3-demethylation is mediated by the same form(s) of cytochrome P-450 involved in theophylline demethylation, while a second form(s) of cytochrome P-450 is involved in theobromine 7-demethylation and theophylline 8-hydroxylation. Theophylline 176-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2865110-9 1985 When compared with previous data relating to theophylline, the results suggest that theobromine 3-demethylation is mediated by the same form(s) of cytochrome P-450 involved in theophylline demethylation, while a second form(s) of cytochrome P-450 is involved in theobromine 7-demethylation and theophylline 8-hydroxylation. Theobromine 262-273 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2865110-9 1985 When compared with previous data relating to theophylline, the results suggest that theobromine 3-demethylation is mediated by the same form(s) of cytochrome P-450 involved in theophylline demethylation, while a second form(s) of cytochrome P-450 is involved in theobromine 7-demethylation and theophylline 8-hydroxylation. Theophylline 176-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2865110-10 1985 In addition, since AMMU formation was inhibited by cimetidine and induced by cigarette smoking and sulfinpyrazone, it would appear that the conversion of theobromine to AMMU is also mediated by cytochrome P-450. ammu 19-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-210 2865110-10 1985 In addition, since AMMU formation was inhibited by cimetidine and induced by cigarette smoking and sulfinpyrazone, it would appear that the conversion of theobromine to AMMU is also mediated by cytochrome P-450. Cimetidine 51-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-210 2865110-10 1985 In addition, since AMMU formation was inhibited by cimetidine and induced by cigarette smoking and sulfinpyrazone, it would appear that the conversion of theobromine to AMMU is also mediated by cytochrome P-450. Sulfinpyrazone 99-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-210 2865110-10 1985 In addition, since AMMU formation was inhibited by cimetidine and induced by cigarette smoking and sulfinpyrazone, it would appear that the conversion of theobromine to AMMU is also mediated by cytochrome P-450. Theobromine 154-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-210 2865110-10 1985 In addition, since AMMU formation was inhibited by cimetidine and induced by cigarette smoking and sulfinpyrazone, it would appear that the conversion of theobromine to AMMU is also mediated by cytochrome P-450. ammu 169-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-210 4044832-6 1985 LTB4 omega-hydroxylase activity in isolated PMN membranes was linear with respect to duration of incubation and protein concentration, was maximal at pH 7.4, had a Km for LTB4 of 0.6 microM, and was dependent on oxygen and on reduced pyridine nucleotides (apparent Km for NADPH = 0.5 microM; apparent Km for NADH = 223 microM). Oxygen 212-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-22 4044832-6 1985 LTB4 omega-hydroxylase activity in isolated PMN membranes was linear with respect to duration of incubation and protein concentration, was maximal at pH 7.4, had a Km for LTB4 of 0.6 microM, and was dependent on oxygen and on reduced pyridine nucleotides (apparent Km for NADPH = 0.5 microM; apparent Km for NADH = 223 microM). pyridine nucleotides 234-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-22 4044832-6 1985 LTB4 omega-hydroxylase activity in isolated PMN membranes was linear with respect to duration of incubation and protein concentration, was maximal at pH 7.4, had a Km for LTB4 of 0.6 microM, and was dependent on oxygen and on reduced pyridine nucleotides (apparent Km for NADPH = 0.5 microM; apparent Km for NADH = 223 microM). NAD 308-312 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-22 4044832-7 1985 The LTB4 omega-hydroxylase was inhibited significantly by carbon monoxide, ferricytochrome c, SKF-525A, and Triton X-100, but was not affected by alpha-naphthoflavone, azide, cyanide, catalase, and superoxide dismutase. Carbon Monoxide 58-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-26 4044832-7 1985 The LTB4 omega-hydroxylase was inhibited significantly by carbon monoxide, ferricytochrome c, SKF-525A, and Triton X-100, but was not affected by alpha-naphthoflavone, azide, cyanide, catalase, and superoxide dismutase. Carbon 48-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-26 4044832-7 1985 The LTB4 omega-hydroxylase was inhibited significantly by carbon monoxide, ferricytochrome c, SKF-525A, and Triton X-100, but was not affected by alpha-naphthoflavone, azide, cyanide, catalase, and superoxide dismutase. Proadifen 94-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-26 4044832-7 1985 The LTB4 omega-hydroxylase was inhibited significantly by carbon monoxide, ferricytochrome c, SKF-525A, and Triton X-100, but was not affected by alpha-naphthoflavone, azide, cyanide, catalase, and superoxide dismutase. Octoxynol 108-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-26 3898085-2 1985 We carried out immunoblot analyses of liver microsomes isolated from eight patients and found that each contained a cytochrome P-450, termed HLp, that reacted with antibodies directed against P-450p, a rat liver cytochrome that is inducible by the anti-glucocorticoid pregnenolone-16 alpha-carbonitrile, by glucocorticoids, by anti-seizure drugs, and by such macrolide antibiotics as triacetyloleandomycin. macrolide antibiotics 359-380 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 3898085-2 1985 We carried out immunoblot analyses of liver microsomes isolated from eight patients and found that each contained a cytochrome P-450, termed HLp, that reacted with antibodies directed against P-450p, a rat liver cytochrome that is inducible by the anti-glucocorticoid pregnenolone-16 alpha-carbonitrile, by glucocorticoids, by anti-seizure drugs, and by such macrolide antibiotics as triacetyloleandomycin. Troleandomycin 384-405 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 3780149-9 1986 In patients who are receiving phenytoin, phenobarbital, or other drugs metabolized by the cytochrome P-450 system, serum concentrations of these drugs may increase as a result of influenza vaccination, and dosage adjustments may be necessary. Phenytoin 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 3780149-9 1986 In patients who are receiving phenytoin, phenobarbital, or other drugs metabolized by the cytochrome P-450 system, serum concentrations of these drugs may increase as a result of influenza vaccination, and dosage adjustments may be necessary. Phenobarbital 41-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 3898085-2 1985 We carried out immunoblot analyses of liver microsomes isolated from eight patients and found that each contained a cytochrome P-450, termed HLp, that reacted with antibodies directed against P-450p, a rat liver cytochrome that is inducible by the anti-glucocorticoid pregnenolone-16 alpha-carbonitrile, by glucocorticoids, by anti-seizure drugs, and by such macrolide antibiotics as triacetyloleandomycin. Pregnenolone Carbonitrile 268-302 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 4026319-2 1985 The cytochrome P-450 preparation was electrophoretically homogeneous and had an apparent monomeric molecular weight of 51,000 as judged by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Sodium Dodecyl Sulfate 139-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 4026319-2 1985 The cytochrome P-450 preparation was electrophoretically homogeneous and had an apparent monomeric molecular weight of 51,000 as judged by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. polyacrylamide 162-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 3935180-1 1985 Emulgen 913, Triton N-101 and sodium cholate were compared for their reconstituting action on the dimethylaniline N-demethylation system containing cytochrome P-450 and NADPH-cytochrome P-450 reductase. Triton N 101 13-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 4016117-3 1985 The compounds inhibit oxidation and binding of cytochrome P-450 substrates of type I (naphthalene, aminopyrine) and of type II (aniline). naphthalene 86-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 4016117-3 1985 The compounds inhibit oxidation and binding of cytochrome P-450 substrates of type I (naphthalene, aminopyrine) and of type II (aniline). Aminopyrine 99-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 4016117-3 1985 The compounds inhibit oxidation and binding of cytochrome P-450 substrates of type I (naphthalene, aminopyrine) and of type II (aniline). aniline 128-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 4016117-8 1985 The data obtained make it possible to estimate the distance (r) between the membranes surface and Fe3+ in the active centre of the enzyme (r less than or equal to 20 A) under the assumption that all molecules of cytochrome P-450 are equally remote from the membrane surface. ferric sulfate 98-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 212-228 4017418-6 1985 In view of the known inhibitory effects of cimetidine on cytochrome P-450-dependent enzymes, our data suggest that this enzyme system does not metabolize a significant fraction of ingested ethanol in subjects who have consumed moderate doses of alcohol for several weeks. Cimetidine 43-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 2996790-2 1985 The toxicity of halogenated alkanes originates from their metabolism by cytochrome P-450 which leads to the formation of reactive intermediates. Alkanes 28-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 2996790-8 1985 The importance of local oxygen concentration and structural arrangement of fatty acids around cytochrome P-450 are emphasized. Fatty Acids 75-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 3935180-1 1985 Emulgen 913, Triton N-101 and sodium cholate were compared for their reconstituting action on the dimethylaniline N-demethylation system containing cytochrome P-450 and NADPH-cytochrome P-450 reductase. Sodium Cholate 30-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 3935180-1 1985 Emulgen 913, Triton N-101 and sodium cholate were compared for their reconstituting action on the dimethylaniline N-demethylation system containing cytochrome P-450 and NADPH-cytochrome P-450 reductase. N,N-dimethylaniline 98-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 4005291-1 1985 Using a classical methodology of purification consisting of three chromatographic steps (Octyl-Sepharose, DEAE-cellulose, CM-cellulose) we have purified a new cytochrome P-450 from human liver microsomes. octyl-sepharose CL-4B 89-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-175 4015695-1 1985 Diltiazem and verapamil were found to be inhibitors of the cytochrome P-450-dependent biotransformation of drugs. Diltiazem 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 4015695-1 1985 Diltiazem and verapamil were found to be inhibitors of the cytochrome P-450-dependent biotransformation of drugs. Verapamil 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 2409076-1 1985 Ferric bleomycin was tested for its ability to catalyze a set of six oxidative reactions characteristic of the heme-containing proteins, cytochrome P-450 and chloroperoxidase. iron bleomycin 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-174 4005291-1 1985 Using a classical methodology of purification consisting of three chromatographic steps (Octyl-Sepharose, DEAE-cellulose, CM-cellulose) we have purified a new cytochrome P-450 from human liver microsomes. DEAE-Cellulose 106-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-175 2409076-1 1985 Ferric bleomycin was tested for its ability to catalyze a set of six oxidative reactions characteristic of the heme-containing proteins, cytochrome P-450 and chloroperoxidase. Heme 111-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-174 4005291-1 1985 Using a classical methodology of purification consisting of three chromatographic steps (Octyl-Sepharose, DEAE-cellulose, CM-cellulose) we have purified a new cytochrome P-450 from human liver microsomes. 7,8-diacetoxy-3-(4-nitrophenyl)coumarin 122-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-175 4015675-2 1985 The formation of 1,4-naphthoquinone, the major methanol-soluble product at early time points, showed an almost total dependence on cytochrome P-450, NADPH-cytochrome P-450 reductase and NADPH, and to a lesser extent on dilauroylphosphatidylcholine. 1,2-dilauroylphosphatidylcholine 219-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 4015675-0 1985 Cytochrome P-450 dependent metabolic activation of 1-naphthol to naphthoquinones and covalent binding species. 1-naphthol 51-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2990349-9 1985 Various substrate analogs of camphor induce varying degrees of low-spin to high-spin shift upon binding to ferric cytochrome P-450 (3). Camphor 29-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 4015675-0 1985 Cytochrome P-450 dependent metabolic activation of 1-naphthol to naphthoquinones and covalent binding species. Naphthoquinones 65-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 4015675-4 1985 1,4-Naphthoquinone formation was dependent on the concentration of both cytochrome P-450 (0.05-0.04 microM) and 1-naphthol (5-50 microM). 1,4-naphthoquinone 0-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 4015675-1 1985 1-Naphthol was metabolised by a fully reconstituted cytochrome P-450 system in the presence of NADPH to methanol-soluble and covalently bound products. 1-naphthol 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 4015675-1 1985 1-Naphthol was metabolised by a fully reconstituted cytochrome P-450 system in the presence of NADPH to methanol-soluble and covalently bound products. Methanol 104-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 4015675-2 1985 The formation of 1,4-naphthoquinone, the major methanol-soluble product at early time points, showed an almost total dependence on cytochrome P-450, NADPH-cytochrome P-450 reductase and NADPH, and to a lesser extent on dilauroylphosphatidylcholine. 1,4-naphthoquinone 17-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 4015675-2 1985 The formation of 1,4-naphthoquinone, the major methanol-soluble product at early time points, showed an almost total dependence on cytochrome P-450, NADPH-cytochrome P-450 reductase and NADPH, and to a lesser extent on dilauroylphosphatidylcholine. Methanol 47-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 4040350-5 1985 Conversely, the treatment of rats with human chorionic gonadotropin (hCG) fully reversed the effect of cis-platinum on heme and cytochrome P-450 concentrations. Cisplatin 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 4015675-6 1985 In the absence of NADPH and NADPH-cytochrome P-450 reductase, 1-naphthol was metabolised, in a cumene hydroperoxide- and cytochrome P-450-dependent reaction, to 1,2- and 1,4-naphthoquinone and covalently bound products. 1-naphthol 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 4015675-6 1985 In the absence of NADPH and NADPH-cytochrome P-450 reductase, 1-naphthol was metabolised, in a cumene hydroperoxide- and cytochrome P-450-dependent reaction, to 1,2- and 1,4-naphthoquinone and covalently bound products. cumene hydroperoxide 95-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 4015675-6 1985 In the absence of NADPH and NADPH-cytochrome P-450 reductase, 1-naphthol was metabolised, in a cumene hydroperoxide- and cytochrome P-450-dependent reaction, to 1,2- and 1,4-naphthoquinone and covalently bound products. 1,4-naphthoquinone 170-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 4015675-8 1985 These data show that cytochrome P-450 catalyses the activation of 1-naphthol to naphthoquinone metabolites and covalently bound species, the latter most likely being derived from naphthoquinones. 1-naphthol 66-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 4015675-8 1985 These data show that cytochrome P-450 catalyses the activation of 1-naphthol to naphthoquinone metabolites and covalently bound species, the latter most likely being derived from naphthoquinones. Naphthoquinones 80-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 4027138-0 1985 Phenacetin O-deethylase: an activity of a cytochrome P-450 showing genetic linkage with that catalysing the 4-hydroxylation of debrisoquine? Debrisoquin 127-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 4015675-8 1985 These data show that cytochrome P-450 catalyses the activation of 1-naphthol to naphthoquinone metabolites and covalently bound species, the latter most likely being derived from naphthoquinones. Naphthoquinones 179-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 4018075-4 1985 The simple first-order kinetics of cytochrome P-450 reduction had been observed in the presence of non-ionic detergent Triton N-101. Triton N 101 119-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 4028286-0 1985 Spin state control of cytochrome P-450 reduction and catalytic activity in a reconstituted P-450 LM2 system as induced by a series of benzphetamine analogues. Benzphetamine 134-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 4028286-1 1985 Reconstituted liposomal cytochrome P-450 LM2 was reacted with a series of benzphetamine analogues as substrates. Benzphetamine 74-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 4018075-5 1985 The apparent biphasity of cytochrome P-450 reduction by dithionite should be the result asymmetric distribution of hemoprotein in microsomal and proteoliposomal membranes as well as in cytochrome oligomers in solution. Dithionite 56-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 3877043-2 1985 In rats and in humans, troleandomycin, erythromycin and erythromycin derivatives induce microsomal enzymes; the induced isozymes of cytochrome P-450 have a high activity for these macrolides but a poor activity with several other substrates. Troleandomycin 23-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 3877043-2 1985 In rats and in humans, troleandomycin, erythromycin and erythromycin derivatives induce microsomal enzymes; the induced isozymes of cytochrome P-450 have a high activity for these macrolides but a poor activity with several other substrates. Erythromycin 39-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 3877043-2 1985 In rats and in humans, troleandomycin, erythromycin and erythromycin derivatives induce microsomal enzymes; the induced isozymes of cytochrome P-450 have a high activity for these macrolides but a poor activity with several other substrates. Erythromycin 56-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 3877043-3 1985 These isozymes actively demethylate and oxidize these macrolides into nitrosoalkanes which form stable, inactive complexes with the iron of cytochrome P-450. demethylate 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 3877043-3 1985 These isozymes actively demethylate and oxidize these macrolides into nitrosoalkanes which form stable, inactive complexes with the iron of cytochrome P-450. Macrolides 54-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 3877043-3 1985 These isozymes actively demethylate and oxidize these macrolides into nitrosoalkanes which form stable, inactive complexes with the iron of cytochrome P-450. nitrosoalkanes 70-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 3877043-3 1985 These isozymes actively demethylate and oxidize these macrolides into nitrosoalkanes which form stable, inactive complexes with the iron of cytochrome P-450. Iron 132-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 3864428-4 1985 Along with previously published information the data from this subject suggest that tolbutamide hydroxylation, debrisoquine hydroxylation, theophylline N-demethylation, and theophylline 8-hydroxylation involve four distinct isozymes of cytochrome P-450. Tolbutamide 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 236-252 2990535-8 1985 Fluorescence lifetime measurements of DPH indicate an average distance of greater than or equal to 60 A between the heme of cytochrome P-450 and DPH. Diphenylhexatriene 38-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-148 2990535-8 1985 Fluorescence lifetime measurements of DPH indicate an average distance of greater than or equal to 60 A between the heme of cytochrome P-450 and DPH. Heme 116-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-148 3875625-1 1985 High-performance liquid chromatographic techniques were developed for the simultaneous detection of metabolites in a cytochrome P-450 model system composed of NADH, haemoglobin and methylene blue. NAD 159-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 3864428-4 1985 Along with previously published information the data from this subject suggest that tolbutamide hydroxylation, debrisoquine hydroxylation, theophylline N-demethylation, and theophylline 8-hydroxylation involve four distinct isozymes of cytochrome P-450. Debrisoquin 111-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 236-252 3864428-4 1985 Along with previously published information the data from this subject suggest that tolbutamide hydroxylation, debrisoquine hydroxylation, theophylline N-demethylation, and theophylline 8-hydroxylation involve four distinct isozymes of cytochrome P-450. Theophylline 139-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 236-252 3864428-4 1985 Along with previously published information the data from this subject suggest that tolbutamide hydroxylation, debrisoquine hydroxylation, theophylline N-demethylation, and theophylline 8-hydroxylation involve four distinct isozymes of cytochrome P-450. Theophylline 173-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 236-252 4039724-0 1985 Inhibition of testicular cytochrome P-450-dependent steroid biosynthesis by cis-platinum. Steroids 52-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 3873598-1 1985 Cytochrome P-450-dependent aryl hydrocarbon hydroxylase (AHH) and 7-ethoxycoumarin O-deethylase activities of a cloned line of human lymphoblastoid AHH-1 cells are inhibited by a monoclonal antibody (MAb 1-7-1) prepared to a 3-methylcholanthrene-induced rat liver cytochrome P-450. Methylcholanthrene 225-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 3873598-2 1985 The monoclonal antibody inhibition determined that a single MAb 1-7-1-sensitive type of cytochrome P-450 is responsible for all of AHH expression in both the basal and benz[a]anthracene-induced cells. anthracene 175-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 4039724-0 1985 Inhibition of testicular cytochrome P-450-dependent steroid biosynthesis by cis-platinum. Cisplatin 76-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 4039724-5 1985 The effect of cis-platinum on the testicular cytochrome P-450 appeared unrelated to deficiencies in heme metabolic processes, in so far that neither was the activity of delta-aminolevulinate synthetase decreased, nor was that of heme oxygenase increased. Cisplatin 14-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 4039724-10 1985 It is suggested that the mechanism of depletive action of cis-platinum on microsomal cytochrome P-450 involves an impairment of the effective assembly of heme and apoprotein moieties. Cisplatin 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 4039724-10 1985 It is suggested that the mechanism of depletive action of cis-platinum on microsomal cytochrome P-450 involves an impairment of the effective assembly of heme and apoprotein moieties. Heme 154-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 4025815-5 1985 The Km and Vmax rates obtained with a human liver microsomal preparation were 0.1 mM and 0.23 nmol 4-hydroxymephenytoin formed/min/nmol cytochrome P-450, respectively. 4-hydroxymephenytoin 99-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-152 2580351-5 1985 The amount of this cytochrome P-450 in human peripheral lymphocytes increased after treatment of the mitogenized lymphocytes with the cytochrome P-450 inducer benz[a]anthracene. anthracene 166-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 4025815-7 1985 Activities varied from 5.6 to 156 pmol 4-hydroxymephenytoin formed/min/nmol cytochrome P-450 in 11 human liver microsomal preparations. 4-hydroxymephenytoin 39-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 3987629-8 1985 These results indicate that the antibodies inhibit side-chain cleavage by binding to a region close to the iron-sulfur protein-binding site, thereby preventing transfer of reducing electrons to the cytochrome P-450. Iron 107-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-214 3987629-8 1985 These results indicate that the antibodies inhibit side-chain cleavage by binding to a region close to the iron-sulfur protein-binding site, thereby preventing transfer of reducing electrons to the cytochrome P-450. Sulfur 112-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-214 3875933-0 1985 Characterization of pirenzepine interaction with cytochrome P-450. Pirenzepine 20-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 3892954-2 1985 We than discuss some evidences for the possible involvement of cytochrome P-450 in the processing of antigens (parallelism between carcinogenesis and immunogenicity induced by aromatic hydrocarbons, high inducibility of cytochrome P-450 dependent enzyme activity in human monocytes, significance of compounds with quinone structure as allergens and carcinogenic agents. Hydrocarbons, Aromatic 176-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 4053242-0 1985 Oxygen activation and olefin oxygenation by iron(III)porphyrin as a model of cytochrome P-450. Oxygen 0-6 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 4053242-0 1985 Oxygen activation and olefin oxygenation by iron(III)porphyrin as a model of cytochrome P-450. Alkenes 22-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 4053242-0 1985 Oxygen activation and olefin oxygenation by iron(III)porphyrin as a model of cytochrome P-450. iron(iii)porphyrin 44-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 2580351-5 1985 The amount of this cytochrome P-450 in human peripheral lymphocytes increased after treatment of the mitogenized lymphocytes with the cytochrome P-450 inducer benz[a]anthracene. anthracene 166-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 3892954-2 1985 We than discuss some evidences for the possible involvement of cytochrome P-450 in the processing of antigens (parallelism between carcinogenesis and immunogenicity induced by aromatic hydrocarbons, high inducibility of cytochrome P-450 dependent enzyme activity in human monocytes, significance of compounds with quinone structure as allergens and carcinogenic agents. quinone 314-321 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 3892954-2 1985 We than discuss some evidences for the possible involvement of cytochrome P-450 in the processing of antigens (parallelism between carcinogenesis and immunogenicity induced by aromatic hydrocarbons, high inducibility of cytochrome P-450 dependent enzyme activity in human monocytes, significance of compounds with quinone structure as allergens and carcinogenic agents. quinone 314-321 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 220-236 3920211-3 1985 Upon formation of the ternary complex between cytochromes P-450, b5, and benzphetamine the percentage of cytochrome P-450 in the high spin state was increased from 28 to 74 (RLM5) and from 9 to 85 (LM2). Benzphetamine 73-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 4004771-1 1985 It is well established that liver microsomal cytochrome P-450 participates in steroid metabolism and probably also in the metabolism of anti-oestrogens such as tamoxifen (Nolvadex). Steroids 78-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 4004771-1 1985 It is well established that liver microsomal cytochrome P-450 participates in steroid metabolism and probably also in the metabolism of anti-oestrogens such as tamoxifen (Nolvadex). Tamoxifen 160-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 4004771-1 1985 It is well established that liver microsomal cytochrome P-450 participates in steroid metabolism and probably also in the metabolism of anti-oestrogens such as tamoxifen (Nolvadex). Tamoxifen 171-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 4004771-3 1985 Therefore a simple sensitive spectrophotometric assay for determining levels of cytochrome P-450-dependent cyclohexane hydroxylation activity in breast and uterine microsomes (microsomal fractions) has been developed. Cyclohexane 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 4004771-4 1985 Cyclohexane was chosen as a substrate because of the relatively high levels of cyclohexane hydroxylase activity in tumour microsomes and because cyclohexane serves as a substrate for several forms of cytochrome P-450. Cyclohexane 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-216 2985426-3 1985 CO, which inhibits the cytochrome P-450-dependent oxygen radical formation, had no effect on the oxidation reaction, suggesting that the source of the reactive oxygen species is not the microsomal mixed-function oxidase. Oxygen 50-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 3921030-7 1985 Spectrophotometric studies revealed that miconazole bound to the cytochrome P-450 component of the placental microsomal aromatase complex and had negligible effect on NADPH-cytochrome c (P-450) reductase activity. Miconazole 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 3986002-0 1985 Altered induction response of hepatic cytochrome P-450 to phenobarbital, 3-methylcholanthrene, and beta-naphthoflavone in organotin-treated animals. Phenobarbital 58-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 3986002-0 1985 Altered induction response of hepatic cytochrome P-450 to phenobarbital, 3-methylcholanthrene, and beta-naphthoflavone in organotin-treated animals. Methylcholanthrene 73-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 3986002-0 1985 Altered induction response of hepatic cytochrome P-450 to phenobarbital, 3-methylcholanthrene, and beta-naphthoflavone in organotin-treated animals. beta-Naphthoflavone 99-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 3986002-1 1985 The effects of tricyclohexyltin hydroxide on the induction of cytochrome P-450 in liver by phenobarbital, 3-methylcholanthrene and beta-naphthoflavone were studied. cyhexatin 15-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3986002-1 1985 The effects of tricyclohexyltin hydroxide on the induction of cytochrome P-450 in liver by phenobarbital, 3-methylcholanthrene and beta-naphthoflavone were studied. Phenobarbital 91-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3986002-1 1985 The effects of tricyclohexyltin hydroxide on the induction of cytochrome P-450 in liver by phenobarbital, 3-methylcholanthrene and beta-naphthoflavone were studied. Methylcholanthrene 106-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3986002-1 1985 The effects of tricyclohexyltin hydroxide on the induction of cytochrome P-450 in liver by phenobarbital, 3-methylcholanthrene and beta-naphthoflavone were studied. beta-Naphthoflavone 131-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3986002-2 1985 A single dose of the organotin (15 mg/kg body wt) prevented the full extent of phenobarbital induction of cytochrome P-450 from occurring; this was the case whether tricyclohexyltin was given 48 hr preceeding a single injection of phenobarbital, or administered simultaneously with the first of three daily doses of the drug. Phenobarbital 79-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 3986002-6 1985 The electrophoretic profiles illustrate clearly that the apoprotein moieties of the various cytochrome P-450 subspecies are affected to a considerable extent by treatment with tricyclohexyltin hydroxide alone, and staining in these bands was noticeably reduced even when phenobarbital was administered together with the organotin. cyhexatin 176-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 3986002-6 1985 The electrophoretic profiles illustrate clearly that the apoprotein moieties of the various cytochrome P-450 subspecies are affected to a considerable extent by treatment with tricyclohexyltin hydroxide alone, and staining in these bands was noticeably reduced even when phenobarbital was administered together with the organotin. Phenobarbital 271-284 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 2983957-0 1985 Inhibition of cytochrome P-450-dependent mixed function oxidation by ethanol. Ethanol 69-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 2983957-1 1985 The mechanism of inhibition of cytochrome P-450-dependent mixed function oxidation by ethanol was studied. Ethanol 86-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 2983957-2 1985 Ethanol competitively inhibited the binding of hexobarbital to liver microsomes, and increased the low spin signal of cytochrome P-450 in the electron spin resonance spectra. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 2983957-3 1985 Therefore, ethanol decreased the substrates bound to ferric cytochrome P-450 in the first step of mixed function oxidation. Ethanol 11-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 2983957-5 1985 The NADPH-dependent reduction of liver microsomal cytochrome P-450 was biphasic and composed of two first-order reactions. NADP 4-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 2983957-6 1985 Ethanol decreased the rate constants of the fast and slow phases of microsomal cytochrome P-450 reduction. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 2983957-7 1985 Thus, it is concluded that the inhibition of drug oxidation by ethanol may be due to the displacement of substrates from cytochrome P-450 and to the inhibition of reduction of cytochrome P-450 by NADPH-cytochrome P-450 reductase. Ethanol 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 2983957-7 1985 Thus, it is concluded that the inhibition of drug oxidation by ethanol may be due to the displacement of substrates from cytochrome P-450 and to the inhibition of reduction of cytochrome P-450 by NADPH-cytochrome P-450 reductase. Ethanol 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-192 3921030-8 1985 These results strongly support direct interaction of miconazole with microsomal cytochrome P-450 in human placental microsomes with high affinity resulting in the inhibition of aromatase activity. Miconazole 53-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 3972828-0 1985 Branchpoint for heme alkylation and metabolite formation in the oxidation of arylacetylenes by cytochrome P-450. Heme 16-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 3972828-0 1985 Branchpoint for heme alkylation and metabolite formation in the oxidation of arylacetylenes by cytochrome P-450. arylacetylenes 77-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 3972828-1 1985 Phenylacetylene and biphenylacetylene are oxidized by cytochrome P-450 to the corresponding arylacetic acids. phenylacetylene 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 3972828-1 1985 Phenylacetylene and biphenylacetylene are oxidized by cytochrome P-450 to the corresponding arylacetic acids. biphenylacetylene 20-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 3972828-1 1985 Phenylacetylene and biphenylacetylene are oxidized by cytochrome P-450 to the corresponding arylacetic acids. arylacetic acids 92-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 3972828-4 1985 In contrast, the inactivation of cytochrome P-450 that occurs during the oxidation of phenylacetylene is insensitive to deuterium substitution. phenylacetylene 86-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 2990136-6 1985 This blockade is due to the interaction of the imidazole structure with the cytochrome p-450. imidazole 47-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 3974403-5 1985 Iproniazid must be directly metabolized by cytochrome P-450 to yield propane and propylene, presumably via an azo ester intermediate which could give rise to an isopropyl radical, the chemical species presumed to be responsible for the hepatoxicity apparent after administration of large doses of iproniazid in vivo. Iproniazid 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 3974403-5 1985 Iproniazid must be directly metabolized by cytochrome P-450 to yield propane and propylene, presumably via an azo ester intermediate which could give rise to an isopropyl radical, the chemical species presumed to be responsible for the hepatoxicity apparent after administration of large doses of iproniazid in vivo. Propane 69-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 3974403-5 1985 Iproniazid must be directly metabolized by cytochrome P-450 to yield propane and propylene, presumably via an azo ester intermediate which could give rise to an isopropyl radical, the chemical species presumed to be responsible for the hepatoxicity apparent after administration of large doses of iproniazid in vivo. propylene 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 3974403-5 1985 Iproniazid must be directly metabolized by cytochrome P-450 to yield propane and propylene, presumably via an azo ester intermediate which could give rise to an isopropyl radical, the chemical species presumed to be responsible for the hepatoxicity apparent after administration of large doses of iproniazid in vivo. Isopropyl radical 161-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 3974403-5 1985 Iproniazid must be directly metabolized by cytochrome P-450 to yield propane and propylene, presumably via an azo ester intermediate which could give rise to an isopropyl radical, the chemical species presumed to be responsible for the hepatoxicity apparent after administration of large doses of iproniazid in vivo. Iproniazid 297-307 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2990136-7 1985 Ascorbinic acid is able to overcome this blockade, indicating that cytochrome p-450 is involved in the ascorbinic acid metabolism which is obligatory for the hydroxylation of cholesterol. ascorbinic acid 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 2990136-7 1985 Ascorbinic acid is able to overcome this blockade, indicating that cytochrome p-450 is involved in the ascorbinic acid metabolism which is obligatory for the hydroxylation of cholesterol. ascorbinic acid 103-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 2990136-7 1985 Ascorbinic acid is able to overcome this blockade, indicating that cytochrome p-450 is involved in the ascorbinic acid metabolism which is obligatory for the hydroxylation of cholesterol. Cholesterol 175-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 3911848-0 1985 Biochemical properties of cytochrome P-450 in relation to steroid oxygenation. Steroids 58-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 3920135-7 1985 Pretreatment of rats with CoCl2, which reduces cytochrome P-450 content, decreased the PBT by 40% and the ABT by 84%. cobaltous chloride 26-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 2579331-0 1985 Measurement of cytochrome P-450 dependent dealkylation of alkoxyphenoxazones in hepatic S9s and hepatocyte homogenates: effects of dicumarol. alkoxyphenoxazones 58-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 3978755-4 1985 The characteristics of the reactions leading to the covalent binding of the N-methyl group of the azo derivative to microsomal protein and its metabolism to form the hydrocarbon, methane, possessed a number of similarities in the apparent kinetic parameters (Km and Vmax), induction, and inhibition patterns indicating a common pathway of metabolism to form a reactive intermediate and the involvement of cytochrome P-450. anthrone 98-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 405-421 3978755-4 1985 The characteristics of the reactions leading to the covalent binding of the N-methyl group of the azo derivative to microsomal protein and its metabolism to form the hydrocarbon, methane, possessed a number of similarities in the apparent kinetic parameters (Km and Vmax), induction, and inhibition patterns indicating a common pathway of metabolism to form a reactive intermediate and the involvement of cytochrome P-450. Hydrocarbons 166-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 405-421 3978755-4 1985 The characteristics of the reactions leading to the covalent binding of the N-methyl group of the azo derivative to microsomal protein and its metabolism to form the hydrocarbon, methane, possessed a number of similarities in the apparent kinetic parameters (Km and Vmax), induction, and inhibition patterns indicating a common pathway of metabolism to form a reactive intermediate and the involvement of cytochrome P-450. Methane 179-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 405-421 2983185-2 1985 Reaction thermodynamics have been calculated for an oxene model for cytochrome P-450 oxidations of four related arylamines: aniline, p-hydroxyaniline, acetanilide, and acetaminophen, by both radical and nonradical mechanisms, using a semiempirical molecular orbital method (modified neglect of differential overlap). oxirene 52-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2983185-2 1985 Reaction thermodynamics have been calculated for an oxene model for cytochrome P-450 oxidations of four related arylamines: aniline, p-hydroxyaniline, acetanilide, and acetaminophen, by both radical and nonradical mechanisms, using a semiempirical molecular orbital method (modified neglect of differential overlap). aniline 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2983185-2 1985 Reaction thermodynamics have been calculated for an oxene model for cytochrome P-450 oxidations of four related arylamines: aniline, p-hydroxyaniline, acetanilide, and acetaminophen, by both radical and nonradical mechanisms, using a semiempirical molecular orbital method (modified neglect of differential overlap). aniline 124-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2983185-2 1985 Reaction thermodynamics have been calculated for an oxene model for cytochrome P-450 oxidations of four related arylamines: aniline, p-hydroxyaniline, acetanilide, and acetaminophen, by both radical and nonradical mechanisms, using a semiempirical molecular orbital method (modified neglect of differential overlap). 4-aminophenol 133-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2983185-2 1985 Reaction thermodynamics have been calculated for an oxene model for cytochrome P-450 oxidations of four related arylamines: aniline, p-hydroxyaniline, acetanilide, and acetaminophen, by both radical and nonradical mechanisms, using a semiempirical molecular orbital method (modified neglect of differential overlap). acetanilide 151-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2983185-2 1985 Reaction thermodynamics have been calculated for an oxene model for cytochrome P-450 oxidations of four related arylamines: aniline, p-hydroxyaniline, acetanilide, and acetaminophen, by both radical and nonradical mechanisms, using a semiempirical molecular orbital method (modified neglect of differential overlap). Acetaminophen 168-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 3838470-10 1985 The scope of the biological activity of Zn-protoporphyrin to alter haem-dependent processes appeared limited in nature, insofar as the microsomal contents of cytochrome P-450 and b5, as well as the aniline hydroxylase, were similar to those of the control animals. zn-protoporphyrin 40-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-181 3856261-1 1985 A previously reported cDNA clone [pP450(1)] coding for a phenobarbital-inducible cytochrome P-450 variant of rat liver microsomal membranes, designated P-450e(U.C. Phenobarbital 57-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 3985310-6 1985 Cyclohexane was chosen as a substrate because of the relatively high levels of cyclohexane hydroxylase activity in tumor microsomes and because cyclohexane serves as a substrate for several forms of cytochrome P-450. Cyclohexane 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 199-215 3938271-0 1985 Inhalation of butanols: changes in the cytochrome P-450 enzyme system. Butanols 14-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 2983784-3 1985 The radicals were shown to change the optical spectra of Fe3+ located in the active site of the enzyme that are similar to those induced by cytochrome P-450 substrates. ferric sulfate 57-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 3938271-1 1985 After inhalation of different butanol isomers for 3 days (2000 ppm) and 5 days (500 ppm), liver and kidney parameters of the microsomal cytochrome P-450 enzyme system were increased. Butanols 30-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-152 3938271-2 1985 sec-Butanol caused the highest increase in cytochrome P-450 concentration with a 47% rise in the kidneys (500 ppm for 5 days) and 33% in the liver (2000 ppm for 3 days). 2-butanol 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 3938271-7 1985 As a conclusion sec-butanol, probably through its metabolite methyl-ethyl-ketone, is the most potent inducer of microsomal cytochrome P-450 in liver and kidney while iso-butanol does not alter total cytochrome P-450. 2-butanol 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 3938271-7 1985 As a conclusion sec-butanol, probably through its metabolite methyl-ethyl-ketone, is the most potent inducer of microsomal cytochrome P-450 in liver and kidney while iso-butanol does not alter total cytochrome P-450. 2-butanol 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 199-215 3938271-7 1985 As a conclusion sec-butanol, probably through its metabolite methyl-ethyl-ketone, is the most potent inducer of microsomal cytochrome P-450 in liver and kidney while iso-butanol does not alter total cytochrome P-450. methylethyl ketone 61-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 3004423-0 1985 Chemiluminescence of luminol caused by interaction of cytochrome P-450 and cytochrome C with cumene hydroperoxide: comparative studies. Luminol 21-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 2983784-5 1985 The alkylating radical substrate analogs covalently bound to microsomal cytochrome P-450 in the vicinity of the active center, resulting in the inhibition of oxidation of type I and II substrates (e. g., aniline and naphthalene). aniline 204-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 3004423-0 1985 Chemiluminescence of luminol caused by interaction of cytochrome P-450 and cytochrome C with cumene hydroperoxide: comparative studies. cumene hydroperoxide 93-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 2983784-5 1985 The alkylating radical substrate analogs covalently bound to microsomal cytochrome P-450 in the vicinity of the active center, resulting in the inhibition of oxidation of type I and II substrates (e. g., aniline and naphthalene). naphthalene 216-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 2983784-6 1985 The value of the spectral binding constant (Ks) for naphthalene in the presence of the radical covalently bound to the cytochrome P-450 active center showed a tendency to increase. naphthalene 52-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 2983784-7 1985 Using the ESR technique, the interaction between Fe3+ and the radical localized in the active site of cytochrome P-450 was demonstrated. ferric sulfate 49-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 2983784-8 1985 The contribution of Fe3+ to the relaxation of the radicals covalently bound to cytochrome P-450 was evaluated from the values of the spin label ESR spectra saturation curves at 77K. ferric sulfate 20-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 3890847-5 1985 The formation of the cytochrome P-450-reductase-complex necessary for oxygen activation by transfer of electrons is dependent on the charge of the phospholipids. Oxygen 70-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 2983785-0 1985 [Effect of phosphatidylglycerol on the incorporation process of cytochrome P-450 in liposomes from dimyristoylphosphatidylcholine]. Phosphatidylglycerols 11-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 3890847-5 1985 The formation of the cytochrome P-450-reductase-complex necessary for oxygen activation by transfer of electrons is dependent on the charge of the phospholipids. Phospholipids 147-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 3966916-5 1985 The competitive inhibition of debrisoquine and bufuralol of each other"s metabolism, together with the similarity in the values for Km and Ki, support the conclusion that the same form of cytochrome P-450 catalyses these two reactions. Debrisoquin 30-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-204 3966916-5 1985 The competitive inhibition of debrisoquine and bufuralol of each other"s metabolism, together with the similarity in the values for Km and Ki, support the conclusion that the same form of cytochrome P-450 catalyses these two reactions. bufuralol 47-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-204 3978151-0 1985 [Effect of chemical modification of tyrosine residues of cholesterol-hydroxylating cytochrome P-450 on the interaction with high-spin effectors]. Tyrosine 36-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 3978151-0 1985 [Effect of chemical modification of tyrosine residues of cholesterol-hydroxylating cytochrome P-450 on the interaction with high-spin effectors]. Cholesterol 57-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 2983785-0 1985 [Effect of phosphatidylglycerol on the incorporation process of cytochrome P-450 in liposomes from dimyristoylphosphatidylcholine]. Dimyristoylphosphatidylcholine 99-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 2983785-1 1985 Using the electron paramagnetic resonance (EPR) method with spin-labeled fatty acids and gel-penetrating chromatography, the effect of phosphatidylglycerol on cytochrome P-450 incorporation into liposomes from dimyristoylphosphatidylcholine was investigated. Phosphatidylglycerols 135-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-175 2983785-1 1985 Using the electron paramagnetic resonance (EPR) method with spin-labeled fatty acids and gel-penetrating chromatography, the effect of phosphatidylglycerol on cytochrome P-450 incorporation into liposomes from dimyristoylphosphatidylcholine was investigated. Dimyristoylphosphatidylcholine 210-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-175 3914938-4 1985 Primary metabolism involves hydroxylation of the alicyclic rings at several carbon atoms by cytochrome P-450-mediated monooxygenase. Carbon 76-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 3986004-0 1985 [Localization of the adrenodoxin-binding fragment of cholesterol hydroxylating cytochrome P-450 from adrenal cortex mitochondria]. Cholesterol 53-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 3914938-9 1985 Covalent modification of tissue macromolecules by reactive intermediates can be responsible for suicide inactivation of cytochrome P-450 and can possibly mediate some long-term toxic effects of PCP. Phencyclidine 194-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 4064901-0 1985 [Primary structure of cholesterol-hydroxylating cytochrome P-450 from the mitochondria of the adrenal cortex in the bull]. Cholesterol 22-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 3987778-5 1985 There was a significant correlation (r (Pearson"s product moment correlation coefficient) = 0.91) between the hepatic phospholipid and cytochrome P-450 concentration. Phospholipids 118-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 3881283-11 1985 Phenobarbital and other anticonvulsants are inducers of cytochrome P-450 and the mixed-function oxidase system. Phenobarbital 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 3000845-1 1985 Two isozymes of cytochrome P-450 were partially purified to specific contents of 7.0 and 0.5 nmol/mg of protein, respectively, from placenta of non-smoking women by chromatography on octyl Sepharose, hydroxylapatite, DEAE-cellulose and CM-cellulose. octyl-sepharose CL-4B 183-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 3000845-1 1985 Two isozymes of cytochrome P-450 were partially purified to specific contents of 7.0 and 0.5 nmol/mg of protein, respectively, from placenta of non-smoking women by chromatography on octyl Sepharose, hydroxylapatite, DEAE-cellulose and CM-cellulose. Durapatite 200-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 3000845-1 1985 Two isozymes of cytochrome P-450 were partially purified to specific contents of 7.0 and 0.5 nmol/mg of protein, respectively, from placenta of non-smoking women by chromatography on octyl Sepharose, hydroxylapatite, DEAE-cellulose and CM-cellulose. DEAE-Cellulose 217-231 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 3000845-1 1985 Two isozymes of cytochrome P-450 were partially purified to specific contents of 7.0 and 0.5 nmol/mg of protein, respectively, from placenta of non-smoking women by chromatography on octyl Sepharose, hydroxylapatite, DEAE-cellulose and CM-cellulose. 7,8-diacetoxy-3-(4-nitrophenyl)coumarin 236-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 3000845-2 1985 NADPH-cytochrome P-450 reductase was purified from phenobarbital-induced mouse liver and from human placenta and was combined with cytochrome P-450 and dilauroylphosphatidylcholine to reconstitute the cytochrome P-450 monooxygenase system. Phenobarbital 51-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 3000845-2 1985 NADPH-cytochrome P-450 reductase was purified from phenobarbital-induced mouse liver and from human placenta and was combined with cytochrome P-450 and dilauroylphosphatidylcholine to reconstitute the cytochrome P-450 monooxygenase system. 1,2-dilauroylphosphatidylcholine 152-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 4029252-3 1985 In all patients the oxidative cytochrome P-450 dependent drug metabolism was induced as shown by an increase of antipyrine-clearance from 36.3 +/- 8.8 to 80.6 +/- 20.1 ml/min and an enhanced urinary excretion of 6-beta-hydroxycortisol from 454 +/- 1.99 to 1607 +/- 362 micrograms/24 h. Furthermore, in all 6 patients the serum alkaline phosphatase declined. Antipyrine 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 4029252-3 1985 In all patients the oxidative cytochrome P-450 dependent drug metabolism was induced as shown by an increase of antipyrine-clearance from 36.3 +/- 8.8 to 80.6 +/- 20.1 ml/min and an enhanced urinary excretion of 6-beta-hydroxycortisol from 454 +/- 1.99 to 1607 +/- 362 micrograms/24 h. Furthermore, in all 6 patients the serum alkaline phosphatase declined. 6 beta-hydroxycortisol 212-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 4065184-2 1985 The subjects with a high liver cytochrome P-450, indicating hepatic microsomal enzyme induction, who showed an increase in liver size, had an elevated high-density lipoprotein concentration and high-density lipoprotein cholesterol/total cholesterol ratio, and a reduced low/high-density lipoprotein cholesterol ratio. Cholesterol 219-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 4065184-2 1985 The subjects with a high liver cytochrome P-450, indicating hepatic microsomal enzyme induction, who showed an increase in liver size, had an elevated high-density lipoprotein concentration and high-density lipoprotein cholesterol/total cholesterol ratio, and a reduced low/high-density lipoprotein cholesterol ratio. density lipoprotein cholesterol 199-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 3965304-0 1985 Catalytic activity of the membrane-bound methylcholanthrene-inducible cytochrome P-450. Methylcholanthrene 41-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 3965304-1 1985 The benzopyrene hydroxylase activity of the methylcholanthrene-inducible form of cytochrome P-450 (P-448) has been studied in native and reconstituted liver microsomal membranes. Methylcholanthrene 44-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 3987778-6 1985 The cytochrome P-450 concentration was inversely related (r = -0.74) to the triacylglycerol concentration. Triglycerides 76-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 3987778-7 1985 The positive correlation between hepatic phospholipids and drug-metabolizing enzymes could be interpreted as indicating that in human liver phospholipid and cytochrome P-450 synthesis share common regulators, or that phospholipids are necessary for the maximum rate of cytochrome P-450 synthesis. Phospholipids 41-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 3987778-7 1985 The positive correlation between hepatic phospholipids and drug-metabolizing enzymes could be interpreted as indicating that in human liver phospholipid and cytochrome P-450 synthesis share common regulators, or that phospholipids are necessary for the maximum rate of cytochrome P-450 synthesis. Phospholipids 41-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 269-285 3987778-7 1985 The positive correlation between hepatic phospholipids and drug-metabolizing enzymes could be interpreted as indicating that in human liver phospholipid and cytochrome P-450 synthesis share common regulators, or that phospholipids are necessary for the maximum rate of cytochrome P-450 synthesis. Phospholipids 41-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 3987778-7 1985 The positive correlation between hepatic phospholipids and drug-metabolizing enzymes could be interpreted as indicating that in human liver phospholipid and cytochrome P-450 synthesis share common regulators, or that phospholipids are necessary for the maximum rate of cytochrome P-450 synthesis. Phospholipids 41-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 269-285 4093989-4 1985 Both primary and secondary oxidation of benzene and its metabolites are mediated via cytochrome P-450 in the liver, although the role of secondary metabolism in the bone marrow is not clear. Benzene 40-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 4009422-0 1985 [Steroid 17 alpha-hydroxylase-C17,20 lyase (cytochrome P-450) from porcine adrenocortical microsomes: inhibition of lyase activity by progesterone and inhibition of hydroxylase activity by 17 alpha-hydroxyprogesterone]. 17-alpha-Hydroxyprogesterone 189-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-60 2422643-1 1985 A new method for the removal of the stabilizing substrate, deoxycorticosterone, from adrenal cytochrome P-450(11) beta, has been developed. Desoxycorticosterone 59-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2422643-3 1985 The substrate-free enzyme, obtained in this manner, has all the characteristic spectral properties of low-spin cytochrome P-450(11) beta and may be converted to the high-spin form by the addition of deoxycorticosterone. Desoxycorticosterone 199-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 2422643-5 1985 It does not require the addition of the cofactors for cytochrome P-450-dependant hydroxylation of deoxycorticosterone, small amounts of enzyme may be prepared in a short time and the enzyme preparation is not diluted to any great extent during the process. Desoxycorticosterone 98-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 2865858-3 1985 Since ethanol is metabolized via a cytochrome P-450 dependent biotransformation system (MEOS) in hepatic microsomes, the microsomal enzyme induction in the smooth endoplasmic reticulum has to be considered as an adaptive response. Ethanol 6-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 3991787-1 1985 Caffeine is mainly metabolized by 3-methylcholanthrene-inducible cytochrome P-450 (P-450MC) and noramidopyrine-methanesulfonate sodium (metamizol, Analgin) is mainly metabolized by phenobarbital-inducible cytochrome P-450 (P-450PB). Caffeine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 3991787-1 1985 Caffeine is mainly metabolized by 3-methylcholanthrene-inducible cytochrome P-450 (P-450MC) and noramidopyrine-methanesulfonate sodium (metamizol, Analgin) is mainly metabolized by phenobarbital-inducible cytochrome P-450 (P-450PB). Methylcholanthrene 34-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 3983470-4 1985 Metyrapone inhibited the stimulation process, indicating that cytochrome P-450 function may be involved. Metyrapone 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 3983470-5 1985 Increase of hydrogen peroxide formation occurred at the same (+)-cyanidanol-3 concentrations that were capable to inhibit lipid peroxidation, suggesting that the interference of (+)-cyanidanol-3 with the oxidase function of cytochrome P-450 does not compromise its antioxidative activity towards lipid peroxidation. Hydrogen Peroxide 12-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 224-240 3983470-5 1985 Increase of hydrogen peroxide formation occurred at the same (+)-cyanidanol-3 concentrations that were capable to inhibit lipid peroxidation, suggesting that the interference of (+)-cyanidanol-3 with the oxidase function of cytochrome P-450 does not compromise its antioxidative activity towards lipid peroxidation. Catechin 61-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 224-240 3983470-5 1985 Increase of hydrogen peroxide formation occurred at the same (+)-cyanidanol-3 concentrations that were capable to inhibit lipid peroxidation, suggesting that the interference of (+)-cyanidanol-3 with the oxidase function of cytochrome P-450 does not compromise its antioxidative activity towards lipid peroxidation. Catechin 178-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 224-240 4009422-0 1985 [Steroid 17 alpha-hydroxylase-C17,20 lyase (cytochrome P-450) from porcine adrenocortical microsomes: inhibition of lyase activity by progesterone and inhibition of hydroxylase activity by 17 alpha-hydroxyprogesterone]. Progesterone 134-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-60 6498831-12 1984 N-OH-AAF:7-OH-AAF, and 7-OH-AAF:1-OH-AAF at the low concentration of AAF, and imply that similar forms of cytochrome P-450 produce these metabolites. 1-oh-aaf 32-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 6084519-5 1984 The present findings also-establish the most facile pathways for enzymatic activation of Fe(III)-bleomycin and Cu(II)-bleomycin, provide data concerning the nature of the activated metallobleomycins, and extend the analogy between the chemistry of cytochrome P-450 and bleomycin. fe(iii)-bleomycin 89-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 248-278 6498831-12 1984 N-OH-AAF:7-OH-AAF, and 7-OH-AAF:1-OH-AAF at the low concentration of AAF, and imply that similar forms of cytochrome P-450 produce these metabolites. 2-Acetylaminofluorene 5-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 6594694-0 1984 Characterization of a human liver cytochrome P-450 involved in the oxidation of debrisoquine and other drugs by using antibodies raised to the analogous rat enzyme. Debrisoquin 80-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 6096874-0 1984 Base (O.-2, e-, or OH-)-induced autoxygenation of organic substrates: a model chemical system for cytochrome P-450-catalyzed monoxygenation and dehydrogenation by dioxygen. Oxygen 163-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 6594694-7 1984 The level of translatable mRNA coding for the debrisoquine-hydroxylating cytochrome P-450 was an order of magnitude less in human liver than in rat liver. Debrisoquin 46-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 6208195-0 1984 Cytochrome P-450-dependent oxidation of lanosterol in cholesterol biosynthesis. Lanosterol 40-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6150639-7 1984 Marketed and investigational H2-antagonist drugs differ in their ability to inhibit drug metabolism due to the combined characteristics of cytochrome P-450 binding affinity and therapeutic dosage. Hydrogen 29-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6208195-0 1984 Cytochrome P-450-dependent oxidation of lanosterol in cholesterol biosynthesis. Cholesterol 54-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6208195-2 1984 Electron transfer to rat liver microsomal cytochrome P-450 of 14 alpha-methyl group demethylation of 24,25-dihydrolanosterol (C30-sterol) has been studied with a new radio-high-performance liquid chromatography assay. lanostenol 101-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 6208195-2 1984 Electron transfer to rat liver microsomal cytochrome P-450 of 14 alpha-methyl group demethylation of 24,25-dihydrolanosterol (C30-sterol) has been studied with a new radio-high-performance liquid chromatography assay. c30-sterol 126-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 6208195-3 1984 The monooxygenase is dependent upon NADPH plus oxygen, insensitive to CN-, and sensitive to CO. Microsomal oxidation is also sensitive to trypsin digestion, and reactivation is dependent upon the addition of purified, detergent-solubilized cytochrome P-450 reductase. Oxygen 8-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-256 6208195-10 1984 Alternatively, administration of complete adjuvant in mineral oil drastically reduces amounts of total microsomal cytochrome P-450 while activity of 14 alpha-methyl sterol oxidase is not affected dramatically. Mineral Oil 54-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 6508979-5 1984 Rifampicin seems to induce preferentially the cytochrome P-450 (iso-) enzyme(s) involved in the demethylation of antipyrine to norantipyrine. Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 6508804-0 1984 Characterization of a common genetic defect of cytochrome P-450 function (debrisoquine-sparteine type polymorphism)--increased Michaelis is Constant (Km) and loss of stereoselectivity of bufuralol 1"-hydroxylation in poor metabolizers. Debrisoquin 74-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 6508979-5 1984 Rifampicin seems to induce preferentially the cytochrome P-450 (iso-) enzyme(s) involved in the demethylation of antipyrine to norantipyrine. Antipyrine 113-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 6508804-0 1984 Characterization of a common genetic defect of cytochrome P-450 function (debrisoquine-sparteine type polymorphism)--increased Michaelis is Constant (Km) and loss of stereoselectivity of bufuralol 1"-hydroxylation in poor metabolizers. Sparteine 87-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 6508804-0 1984 Characterization of a common genetic defect of cytochrome P-450 function (debrisoquine-sparteine type polymorphism)--increased Michaelis is Constant (Km) and loss of stereoselectivity of bufuralol 1"-hydroxylation in poor metabolizers. bufuralol 187-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 6508804-4 1984 In a non-membraneous reconstituted system containing NADPH cytochrome P-450 reductase, a NADPH regenerating system and phospholipids, P-450[buf] exhibited an almost complete substrate stereoselectivity for (+)-isomer 1"-hydroxylation. NADP 53-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 6508804-4 1984 In a non-membraneous reconstituted system containing NADPH cytochrome P-450 reductase, a NADPH regenerating system and phospholipids, P-450[buf] exhibited an almost complete substrate stereoselectivity for (+)-isomer 1"-hydroxylation. Phospholipids 119-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 6508804-5 1984 It is concluded that the purified cytochrome P-450[buf] is the target of the debrisoquine-sparteine type oxidation polymorphism and that poor metabolizers have a quantitative or qualitative deficiency of this isozyme. Debrisoquin 77-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 6508979-5 1984 Rifampicin seems to induce preferentially the cytochrome P-450 (iso-) enzyme(s) involved in the demethylation of antipyrine to norantipyrine. Edaravone 127-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 6508804-5 1984 It is concluded that the purified cytochrome P-450[buf] is the target of the debrisoquine-sparteine type oxidation polymorphism and that poor metabolizers have a quantitative or qualitative deficiency of this isozyme. Sparteine 90-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 6508979-7 1984 This provides further evidence for the involvement of different iso-enzymes of the cytochrome P-450 system in antipyrine metabolism in man. Antipyrine 110-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 6096071-3 1984 Like cimetidine, ranitidine binds to cytochrome P-450 in the liver where it appears to exert an inhibitory effect, but to a lesser extent than cimetidine. Cimetidine 5-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 6525207-0 1984 [Molecular organization of cholesterol-hydroxylating cytochrome P-450 from adrenal cortex mitochondria. Cholesterol 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 6096071-3 1984 Like cimetidine, ranitidine binds to cytochrome P-450 in the liver where it appears to exert an inhibitory effect, but to a lesser extent than cimetidine. Ranitidine 17-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 6096071-13 1984 The reported interactions of ranitidine with warfarin, metoprolol, nifedipine, theophylline and fentanyl appear to be due to inhibition of cytochrome P-450. Ranitidine 29-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6096071-13 1984 The reported interactions of ranitidine with warfarin, metoprolol, nifedipine, theophylline and fentanyl appear to be due to inhibition of cytochrome P-450. Warfarin 45-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6096071-13 1984 The reported interactions of ranitidine with warfarin, metoprolol, nifedipine, theophylline and fentanyl appear to be due to inhibition of cytochrome P-450. Metoprolol 55-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6096071-13 1984 The reported interactions of ranitidine with warfarin, metoprolol, nifedipine, theophylline and fentanyl appear to be due to inhibition of cytochrome P-450. Nifedipine 67-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6096071-13 1984 The reported interactions of ranitidine with warfarin, metoprolol, nifedipine, theophylline and fentanyl appear to be due to inhibition of cytochrome P-450. Theophylline 79-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6096071-13 1984 The reported interactions of ranitidine with warfarin, metoprolol, nifedipine, theophylline and fentanyl appear to be due to inhibition of cytochrome P-450. Fentanyl 96-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6093289-5 1984 The time frame during which enhanced sensitivity to parathion and pentobarbital was observed suggests that this sensitivity may have been caused by viral-induced interferon-mediated depression of cytochrome P-450. Parathion 52-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 196-212 6093289-5 1984 The time frame during which enhanced sensitivity to parathion and pentobarbital was observed suggests that this sensitivity may have been caused by viral-induced interferon-mediated depression of cytochrome P-450. Pentobarbital 66-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 196-212 6435621-2 1984 This MVP analog has been found to be a highly efficient and specific chemiluminescent probe for picomole quantities of singlet oxygen and singlet oxygen equivalents, and it produces significant chemiluminescence when reacted with cytochrome P-450 enzymes. Singlet Oxygen 119-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-246 6091674-0 1984 Increased microsomal oxidation of ethanol by cytochrome P-450 and hydroxyl radical-dependent pathways after chronic ethanol consumption. Ethanol 34-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 6091674-0 1984 Increased microsomal oxidation of ethanol by cytochrome P-450 and hydroxyl radical-dependent pathways after chronic ethanol consumption. Ethanol 116-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 6435533-0 1984 The association of cytochrome P-450 and NADPH-cytochrome P-450 reductase in phospholipid membranes. Phospholipids 76-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 6435533-1 1984 NADPH-cytochrome P-450 reductase and two purified isozymes of cytochrome P-450 have been incorporated into phospholipid vesicles by a cholate dialysis technique. Phospholipids 107-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 6435621-2 1984 This MVP analog has been found to be a highly efficient and specific chemiluminescent probe for picomole quantities of singlet oxygen and singlet oxygen equivalents, and it produces significant chemiluminescence when reacted with cytochrome P-450 enzymes. Singlet Oxygen 138-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-246 6091554-12 1984 This suggestion implies that the electron distribution of the iron at the catalytic sites of cytochrome P-450 and certain chlorin-containing enzymes is in some way similar, but distinct from that at the transport site of myoglobin. Iron 62-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 6477826-1 1984 Arylhydrocarbon-hydroxylase (AHH) is a cytochrome P-450-dependent polysubstrate mono-oxygenase which plays an important role in converting some compounds (e.g. benzo[a]pyrene) to highly reactive carcinogenic species. Benzo(a)pyrene 160-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 6088105-9 1984 These results demonstrate that oxidation products of the peroxidation of lipids and fatty acids are able to react directly with benzo[a]pyrene to form products including benzo[a]pyrene quinones without the presence of enzymes such as the cytochrome P-450 mixed function oxidase system and prostaglandin synthetase. Fatty Acids 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 238-254 6487483-6 1984 Previous studies have suggested that two forms of cytochrome P-450 are involved in theophylline metabolism, one mediating the N-demethylations and the other 8-oxidation. Theophylline 83-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 6508858-0 1984 [Localization of tetranitromethane-modified tyrosine residues in domains of cholesterol-hydroxylating cytochrome P-450]. Tetranitromethane 17-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-119 6508858-0 1984 [Localization of tetranitromethane-modified tyrosine residues in domains of cholesterol-hydroxylating cytochrome P-450]. Tyrosine 44-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-119 6508858-0 1984 [Localization of tetranitromethane-modified tyrosine residues in domains of cholesterol-hydroxylating cytochrome P-450]. Cholesterol 76-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-119 6508858-2 1984 With the aid of thiol-disulfide exchange chromatography and SDS polyacrylamide gel electrophoresis the F1 and F2 fragments of the modified cytochrome P-450 were isolated. Sulfhydryl Compounds 16-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6508858-2 1984 With the aid of thiol-disulfide exchange chromatography and SDS polyacrylamide gel electrophoresis the F1 and F2 fragments of the modified cytochrome P-450 were isolated. Disulfides 22-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6508858-2 1984 With the aid of thiol-disulfide exchange chromatography and SDS polyacrylamide gel electrophoresis the F1 and F2 fragments of the modified cytochrome P-450 were isolated. Sodium Dodecyl Sulfate 60-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6508858-2 1984 With the aid of thiol-disulfide exchange chromatography and SDS polyacrylamide gel electrophoresis the F1 and F2 fragments of the modified cytochrome P-450 were isolated. polyacrylamide 64-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 6088105-9 1984 These results demonstrate that oxidation products of the peroxidation of lipids and fatty acids are able to react directly with benzo[a]pyrene to form products including benzo[a]pyrene quinones without the presence of enzymes such as the cytochrome P-450 mixed function oxidase system and prostaglandin synthetase. Benzo(a)pyrene 128-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 238-254 6471064-0 1984 Oxidation of sparteines by cytochrome P-450: evidence against the formation of N-oxides. Sparteine 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 6438379-0 1984 Cytochrome P-450-dependent oxidative cleavage of 1-(tetrahydro-2-furanyl)-5-fluorouracil to 5-fluorouracil. Tegafur 49-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6438379-0 1984 Cytochrome P-450-dependent oxidative cleavage of 1-(tetrahydro-2-furanyl)-5-fluorouracil to 5-fluorouracil. Fluorouracil 74-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6438379-1 1984 Cytochrome P-450-dependent oxidative cleavage of 1-(tetrahydro-2-furanyl)-5-fluorouracil (FT) was investigated in a reconstituted system containing purified phenobarbital-inducible cytochrome P-450 (P-450(1)) or 3-methylcholanthrene-inducible cytochrome P-450 (P-448(1)). Tegafur 49-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6438379-1 1984 Cytochrome P-450-dependent oxidative cleavage of 1-(tetrahydro-2-furanyl)-5-fluorouracil (FT) was investigated in a reconstituted system containing purified phenobarbital-inducible cytochrome P-450 (P-450(1)) or 3-methylcholanthrene-inducible cytochrome P-450 (P-448(1)). Tegafur 49-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 6438379-1 1984 Cytochrome P-450-dependent oxidative cleavage of 1-(tetrahydro-2-furanyl)-5-fluorouracil (FT) was investigated in a reconstituted system containing purified phenobarbital-inducible cytochrome P-450 (P-450(1)) or 3-methylcholanthrene-inducible cytochrome P-450 (P-448(1)). Tegafur 90-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6438379-1 1984 Cytochrome P-450-dependent oxidative cleavage of 1-(tetrahydro-2-furanyl)-5-fluorouracil (FT) was investigated in a reconstituted system containing purified phenobarbital-inducible cytochrome P-450 (P-450(1)) or 3-methylcholanthrene-inducible cytochrome P-450 (P-448(1)). Tegafur 90-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 6438379-1 1984 Cytochrome P-450-dependent oxidative cleavage of 1-(tetrahydro-2-furanyl)-5-fluorouracil (FT) was investigated in a reconstituted system containing purified phenobarbital-inducible cytochrome P-450 (P-450(1)) or 3-methylcholanthrene-inducible cytochrome P-450 (P-448(1)). Phenobarbital 157-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6438379-1 1984 Cytochrome P-450-dependent oxidative cleavage of 1-(tetrahydro-2-furanyl)-5-fluorouracil (FT) was investigated in a reconstituted system containing purified phenobarbital-inducible cytochrome P-450 (P-450(1)) or 3-methylcholanthrene-inducible cytochrome P-450 (P-448(1)). Methylcholanthrene 212-232 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6431911-0 1984 Studies on cytochrome P-450-dependent lipid hydroperoxide reduction. Lipid Peroxides 38-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 6431911-1 1984 A reconstituted mixed-function oxidase system containing cytochrome P-450, cytochrome P-450 reductase, phosphatidylcholine, and NADPH catalyzed the reduction of 13-hydroperoxy-9,11-octadecadienoic acid to 13-hydroxy-9,11-octadecadienoic acid. 13-hydroperoxy-9,11-octadecadienoic acid 161-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 6431911-1 1984 A reconstituted mixed-function oxidase system containing cytochrome P-450, cytochrome P-450 reductase, phosphatidylcholine, and NADPH catalyzed the reduction of 13-hydroperoxy-9,11-octadecadienoic acid to 13-hydroxy-9,11-octadecadienoic acid. 13-hydroperoxy-9,11-octadecadienoic acid 161-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 6431911-1 1984 A reconstituted mixed-function oxidase system containing cytochrome P-450, cytochrome P-450 reductase, phosphatidylcholine, and NADPH catalyzed the reduction of 13-hydroperoxy-9,11-octadecadienoic acid to 13-hydroxy-9,11-octadecadienoic acid. 13-hydroxy-9,11-octadecadienoic acid 205-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 6431911-1 1984 A reconstituted mixed-function oxidase system containing cytochrome P-450, cytochrome P-450 reductase, phosphatidylcholine, and NADPH catalyzed the reduction of 13-hydroperoxy-9,11-octadecadienoic acid to 13-hydroxy-9,11-octadecadienoic acid. 13-hydroxy-9,11-octadecadienoic acid 205-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 6431911-4 1984 Incubation of cytochrome P-450 with only lipid hydroperoxide resulted in destruction of the hemoprotein. Lipid Peroxides 41-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 6431911-5 1984 Addition of substrates such as aminopyrine decreased cytochrome P-450 destruction. Aminopyrine 31-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 6548701-0 1984 Control of heme and cytochrome P-450 metabolism by inorganic metals, organometals and synthetic metalloporphyrins. Metalloporphyrins 96-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 6747992-0 1984 The preferred solution conformation of warfarin at the active site of cytochrome P-450 based on the CD spectra in octanol/water model system. Warfarin 39-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 6747992-0 1984 The preferred solution conformation of warfarin at the active site of cytochrome P-450 based on the CD spectra in octanol/water model system. Octanols 114-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 6747992-0 1984 The preferred solution conformation of warfarin at the active site of cytochrome P-450 based on the CD spectra in octanol/water model system. Water 122-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 6747992-5 1984 On the basis of these results, the hemiketal form is proposed as the preferred solution conformation of warfarin in the lipid environment of the active site of cytochrome P-450 and the relationship between solution conformation and stereoselectivity of warfarin metabolism by beta-naphthoflavone inducible cytochrome P-450 is discussed. Warfarin 104-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 6747992-5 1984 On the basis of these results, the hemiketal form is proposed as the preferred solution conformation of warfarin in the lipid environment of the active site of cytochrome P-450 and the relationship between solution conformation and stereoselectivity of warfarin metabolism by beta-naphthoflavone inducible cytochrome P-450 is discussed. Warfarin 104-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 306-322 6747992-5 1984 On the basis of these results, the hemiketal form is proposed as the preferred solution conformation of warfarin in the lipid environment of the active site of cytochrome P-450 and the relationship between solution conformation and stereoselectivity of warfarin metabolism by beta-naphthoflavone inducible cytochrome P-450 is discussed. Warfarin 253-261 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 6747992-5 1984 On the basis of these results, the hemiketal form is proposed as the preferred solution conformation of warfarin in the lipid environment of the active site of cytochrome P-450 and the relationship between solution conformation and stereoselectivity of warfarin metabolism by beta-naphthoflavone inducible cytochrome P-450 is discussed. Warfarin 253-261 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 306-322 6747992-5 1984 On the basis of these results, the hemiketal form is proposed as the preferred solution conformation of warfarin in the lipid environment of the active site of cytochrome P-450 and the relationship between solution conformation and stereoselectivity of warfarin metabolism by beta-naphthoflavone inducible cytochrome P-450 is discussed. beta-Naphthoflavone 276-295 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 6747992-5 1984 On the basis of these results, the hemiketal form is proposed as the preferred solution conformation of warfarin in the lipid environment of the active site of cytochrome P-450 and the relationship between solution conformation and stereoselectivity of warfarin metabolism by beta-naphthoflavone inducible cytochrome P-450 is discussed. beta-Naphthoflavone 276-295 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 306-322 6548701-3 1984 The resulting increase in the rate of heme breakdown is reflected in a marked depression of cellular cytochrome P-450 content and impairment of the oxidative metabolism of natural and foreign chemicals dependent on this hemeprotein. Heme 38-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-117 6548701-9 1984 These differences, among others which characterize metal actions in vivo and in vitro attest to the importance of pharmacokinetic, adaptive and other host factors in defining the responses of the heme-cytochrome P-450 systems to the impact of metals in the whole animal. Metals 51-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-217 6744247-3 1984 Additional evidence is provided by the regiochemistry of cytochrome P-450 catalyzed oxidation of quinoline. quinoline 97-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 6484850-3 1984 Placentas from normal infants were found to have high levels of monooxygenase activities and low apparent Kms toward ethoxyresorufin (10(-7) M), reflecting induction of cytochrome P-450 enzymes usually associated with maternal cigarette smoking. ethoxyresorufin 117-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 6429140-1 1984 Phospholipid has been reported to be necessary for optimal catalytic activity of a number of mammalian cytochrome P-450 (P-450) systems. Phospholipids 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 6148215-2 1984 In vitro microsomal metabolism of the R and S enantiomers of warfarin to dehydrowarfarin and 4"-, 6-, 7-, 8-, and 10-hydroxywarfarin is catalyzed by cytochrome P-450 isozymes and was used as the basis for evaluating similarities and differences between human cytochrome P-450 isozyme compositions. Warfarin 61-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 6148207-0 1984 Metabolism of alcohol and ketone by cytochrome P-450 oxygenase: fluoren-9-ol in equilibrium with fluoren-9-one. Alcohols 14-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 6148215-2 1984 In vitro microsomal metabolism of the R and S enantiomers of warfarin to dehydrowarfarin and 4"-, 6-, 7-, 8-, and 10-hydroxywarfarin is catalyzed by cytochrome P-450 isozymes and was used as the basis for evaluating similarities and differences between human cytochrome P-450 isozyme compositions. Warfarin 61-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 259-275 6148207-0 1984 Metabolism of alcohol and ketone by cytochrome P-450 oxygenase: fluoren-9-ol in equilibrium with fluoren-9-one. Ketones 26-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 6148215-2 1984 In vitro microsomal metabolism of the R and S enantiomers of warfarin to dehydrowarfarin and 4"-, 6-, 7-, 8-, and 10-hydroxywarfarin is catalyzed by cytochrome P-450 isozymes and was used as the basis for evaluating similarities and differences between human cytochrome P-450 isozyme compositions. Dehydrowarfarin 73-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 6148207-0 1984 Metabolism of alcohol and ketone by cytochrome P-450 oxygenase: fluoren-9-ol in equilibrium with fluoren-9-one. fluoren-9-ol 64-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 6148215-2 1984 In vitro microsomal metabolism of the R and S enantiomers of warfarin to dehydrowarfarin and 4"-, 6-, 7-, 8-, and 10-hydroxywarfarin is catalyzed by cytochrome P-450 isozymes and was used as the basis for evaluating similarities and differences between human cytochrome P-450 isozyme compositions. Dehydrowarfarin 73-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 259-275 6148215-2 1984 In vitro microsomal metabolism of the R and S enantiomers of warfarin to dehydrowarfarin and 4"-, 6-, 7-, 8-, and 10-hydroxywarfarin is catalyzed by cytochrome P-450 isozymes and was used as the basis for evaluating similarities and differences between human cytochrome P-450 isozyme compositions. 4"-, 6-, 7-, 8-, and 10-hydroxywarfarin 93-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 6148207-0 1984 Metabolism of alcohol and ketone by cytochrome P-450 oxygenase: fluoren-9-ol in equilibrium with fluoren-9-one. 9-fluorenone 97-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 6148215-2 1984 In vitro microsomal metabolism of the R and S enantiomers of warfarin to dehydrowarfarin and 4"-, 6-, 7-, 8-, and 10-hydroxywarfarin is catalyzed by cytochrome P-450 isozymes and was used as the basis for evaluating similarities and differences between human cytochrome P-450 isozyme compositions. 4"-, 6-, 7-, 8-, and 10-hydroxywarfarin 93-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 259-275 6148215-0 1984 Human hepatic cytochrome P-450 composition as probed by in vitro microsomal metabolism of warfarin. Warfarin 90-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 6148215-8 1984 Based on the known warfarin metabolite profiles of five purified cytochrome P-450 isozymes, the isozyme composition of the microsomes can be estimated. Warfarin 19-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 6725517-1 1984 Dietary protein, when substituted for carbohydrate or fat, can increase cytochrome P-450-dependent drug oxidation rates in humans. Carbohydrates 38-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6506763-0 1984 The measurement of FAD-containing mono-oxygenase activity in microsomes containing cytochrome P-450. Flavin-Adenine Dinucleotide 19-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 6611155-1 1984 Deuterium labeling of 7-ethoxycoumarin on the carbon undergoing oxidation by cytochrome P-450 results in a redirection of metabolism from O-deethylation to ring hydroxylation at C6 (Harada, N. et al (1984) J Biol Chem 259, 3005-3010). Deuterium 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 6611155-1 1984 Deuterium labeling of 7-ethoxycoumarin on the carbon undergoing oxidation by cytochrome P-450 results in a redirection of metabolism from O-deethylation to ring hydroxylation at C6 (Harada, N. et al (1984) J Biol Chem 259, 3005-3010). 7-ethoxycoumarin 22-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 6611155-1 1984 Deuterium labeling of 7-ethoxycoumarin on the carbon undergoing oxidation by cytochrome P-450 results in a redirection of metabolism from O-deethylation to ring hydroxylation at C6 (Harada, N. et al (1984) J Biol Chem 259, 3005-3010). Carbon 46-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 6725272-2 1984 This laboratory has recently reported that, in a reconstituted enzyme system containing alcohol-induced isozyme 3a of liver microsomal cytochrome P-450, the sum of acetaldehyde generated by the monooxygenation of ethanol and of hydrogen peroxide produced by the NADPH oxidase activity is inadequate to account for the O2 and NADPH consumed. Alcohols 88-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 6725272-2 1984 This laboratory has recently reported that, in a reconstituted enzyme system containing alcohol-induced isozyme 3a of liver microsomal cytochrome P-450, the sum of acetaldehyde generated by the monooxygenation of ethanol and of hydrogen peroxide produced by the NADPH oxidase activity is inadequate to account for the O2 and NADPH consumed. Acetaldehyde 164-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 6725272-2 1984 This laboratory has recently reported that, in a reconstituted enzyme system containing alcohol-induced isozyme 3a of liver microsomal cytochrome P-450, the sum of acetaldehyde generated by the monooxygenation of ethanol and of hydrogen peroxide produced by the NADPH oxidase activity is inadequate to account for the O2 and NADPH consumed. Ethanol 213-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 6725272-2 1984 This laboratory has recently reported that, in a reconstituted enzyme system containing alcohol-induced isozyme 3a of liver microsomal cytochrome P-450, the sum of acetaldehyde generated by the monooxygenation of ethanol and of hydrogen peroxide produced by the NADPH oxidase activity is inadequate to account for the O2 and NADPH consumed. Hydrogen Peroxide 228-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 6725272-2 1984 This laboratory has recently reported that, in a reconstituted enzyme system containing alcohol-induced isozyme 3a of liver microsomal cytochrome P-450, the sum of acetaldehyde generated by the monooxygenation of ethanol and of hydrogen peroxide produced by the NADPH oxidase activity is inadequate to account for the O2 and NADPH consumed. Oxygen 318-320 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 6725272-2 1984 This laboratory has recently reported that, in a reconstituted enzyme system containing alcohol-induced isozyme 3a of liver microsomal cytochrome P-450, the sum of acetaldehyde generated by the monooxygenation of ethanol and of hydrogen peroxide produced by the NADPH oxidase activity is inadequate to account for the O2 and NADPH consumed. NADP 262-267 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 6088030-1 1984 Using the all-valence electron, semiempirical molecular orbital method, MNDO, properties have been identified and calculated for eight chloroethanes which can serve as indicators of their extent of transformation to alcohols by cytochrome P450 and the subsequent formation of aldehydes by loss of HCl from these alcohols. Ethyl Chloride 135-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 6542186-0 1984 Bioorganotin chemistry: a commentary on the reactions of organotin compounds with a cytochrome P-450 dependent monooxygenase enzyme system. bioorganotin 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 6542186-1 1984 The biological oxidation of several tributyltin derivatives, by a cytochrome P-450 dependent monooxygenase enzyme system with reduced nicotinamideadeninedinucleotidephosphate as the essential cofactor, produced carbon-hydroxylated compounds identified as alpha-, beta-, gamma- and delta-hydroxybutyldibutyltin derivatives. tributyltin 36-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 6542186-1 1984 The biological oxidation of several tributyltin derivatives, by a cytochrome P-450 dependent monooxygenase enzyme system with reduced nicotinamideadeninedinucleotidephosphate as the essential cofactor, produced carbon-hydroxylated compounds identified as alpha-, beta-, gamma- and delta-hydroxybutyldibutyltin derivatives. NADP 134-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 6542186-1 1984 The biological oxidation of several tributyltin derivatives, by a cytochrome P-450 dependent monooxygenase enzyme system with reduced nicotinamideadeninedinucleotidephosphate as the essential cofactor, produced carbon-hydroxylated compounds identified as alpha-, beta-, gamma- and delta-hydroxybutyldibutyltin derivatives. Carbon 211-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 6542186-1 1984 The biological oxidation of several tributyltin derivatives, by a cytochrome P-450 dependent monooxygenase enzyme system with reduced nicotinamideadeninedinucleotidephosphate as the essential cofactor, produced carbon-hydroxylated compounds identified as alpha-, beta-, gamma- and delta-hydroxybutyldibutyltin derivatives. alpha-, beta-, gamma- and delta-hydroxybutyldibutyltin 255-309 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 6088030-1 1984 Using the all-valence electron, semiempirical molecular orbital method, MNDO, properties have been identified and calculated for eight chloroethanes which can serve as indicators of their extent of transformation to alcohols by cytochrome P450 and the subsequent formation of aldehydes by loss of HCl from these alcohols. Alcohols 216-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 6088030-1 1984 Using the all-valence electron, semiempirical molecular orbital method, MNDO, properties have been identified and calculated for eight chloroethanes which can serve as indicators of their extent of transformation to alcohols by cytochrome P450 and the subsequent formation of aldehydes by loss of HCl from these alcohols. Aldehydes 276-285 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 6088030-1 1984 Using the all-valence electron, semiempirical molecular orbital method, MNDO, properties have been identified and calculated for eight chloroethanes which can serve as indicators of their extent of transformation to alcohols by cytochrome P450 and the subsequent formation of aldehydes by loss of HCl from these alcohols. Hydrochloric Acid 297-300 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 6088030-1 1984 Using the all-valence electron, semiempirical molecular orbital method, MNDO, properties have been identified and calculated for eight chloroethanes which can serve as indicators of their extent of transformation to alcohols by cytochrome P450 and the subsequent formation of aldehydes by loss of HCl from these alcohols. Alcohols 312-320 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 6088030-5 1984 The molecular properties identified in this study as indicators of rank order or carcinogenic activity of the parent chloroethanes are consistent with the importance of cytochrome P450 in transforming halohydrocarbons to active carcinogens and of acylchlorides and chloroaldehydes as the active form. Ethyl Chloride 117-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-184 6088030-5 1984 The molecular properties identified in this study as indicators of rank order or carcinogenic activity of the parent chloroethanes are consistent with the importance of cytochrome P450 in transforming halohydrocarbons to active carcinogens and of acylchlorides and chloroaldehydes as the active form. Hydrocarbons, Halogenated 201-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-184 6088030-5 1984 The molecular properties identified in this study as indicators of rank order or carcinogenic activity of the parent chloroethanes are consistent with the importance of cytochrome P450 in transforming halohydrocarbons to active carcinogens and of acylchlorides and chloroaldehydes as the active form. chloroacetaldehyde 265-280 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-184 6704207-1 1984 Cytochrome P-450 and NADPH cytochrome P-450 reductase were incorporated into large unilamellar lipid vesicles (200-300 nm in diameter) removing octylglucoside from mixed micelles by dialysis. octyl-beta-D-glucoside 144-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-53 6706995-1 1984 The terminal acetylenic analogue of lauric acid, 11-dodecynoic acid (11-DDYA), specifically inactivates hepatic cytochrome P-450 enzymes that catalyze omega- and omega-1-hydroxylation of lauric acid. acetylenic 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 6587349-0 1984 Epoxidation of olefins by cytochrome P-450 model compounds: mechanism of oxygen atom transfer. Alkenes 15-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 6587349-0 1984 Epoxidation of olefins by cytochrome P-450 model compounds: mechanism of oxygen atom transfer. Oxygen 73-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 6587349-5 1984 Some analogies to the biochemical epoxidation of olefins catalyzed by cytochrome P-450 are discussed. Alkenes 49-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 6706995-1 1984 The terminal acetylenic analogue of lauric acid, 11-dodecynoic acid (11-DDYA), specifically inactivates hepatic cytochrome P-450 enzymes that catalyze omega- and omega-1-hydroxylation of lauric acid. lauric acid 36-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 6706995-1 1984 The terminal acetylenic analogue of lauric acid, 11-dodecynoic acid (11-DDYA), specifically inactivates hepatic cytochrome P-450 enzymes that catalyze omega- and omega-1-hydroxylation of lauric acid. 11-dodecynoic acid 49-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 6706995-1 1984 The terminal acetylenic analogue of lauric acid, 11-dodecynoic acid (11-DDYA), specifically inactivates hepatic cytochrome P-450 enzymes that catalyze omega- and omega-1-hydroxylation of lauric acid. 11-dodecynoic acid 69-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 6706995-1 1984 The terminal acetylenic analogue of lauric acid, 11-dodecynoic acid (11-DDYA), specifically inactivates hepatic cytochrome P-450 enzymes that catalyze omega- and omega-1-hydroxylation of lauric acid. lauric acid 187-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 6088753-5 1984 The epoxide was found to resist the hydrolysis by epoxide hydrolase, and was further converted to several metabolites by monooxygenase system involving cytochrome P-450. Epoxy Compounds 4-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-168 6200138-4 1984 Scavengers of hydrogen peroxide and hydroxyl radicals, including catalase and benzoate and inhibitors of microsomal cytochrome P-450-dependent monooxygenases such as 1-benzylimidazole, metyrapone and alpha-naphthoflavone, had no inhibitory effects on bleomycin-mediated DNA chain breakage. 1-benzylimidazole 166-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 6200138-4 1984 Scavengers of hydrogen peroxide and hydroxyl radicals, including catalase and benzoate and inhibitors of microsomal cytochrome P-450-dependent monooxygenases such as 1-benzylimidazole, metyrapone and alpha-naphthoflavone, had no inhibitory effects on bleomycin-mediated DNA chain breakage. Metyrapone 185-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 6200138-4 1984 Scavengers of hydrogen peroxide and hydroxyl radicals, including catalase and benzoate and inhibitors of microsomal cytochrome P-450-dependent monooxygenases such as 1-benzylimidazole, metyrapone and alpha-naphthoflavone, had no inhibitory effects on bleomycin-mediated DNA chain breakage. alpha-naphthoflavone 200-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 6200138-4 1984 Scavengers of hydrogen peroxide and hydroxyl radicals, including catalase and benzoate and inhibitors of microsomal cytochrome P-450-dependent monooxygenases such as 1-benzylimidazole, metyrapone and alpha-naphthoflavone, had no inhibitory effects on bleomycin-mediated DNA chain breakage. Bleomycin 251-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 6088753-9 1984 These results indicate that 9 alpha, 10 alpha-EHHC formed from delta 9-THC is further metabolized not by epoxide hydrolase but by monooxygenase system involving cytochrome P-450, and that, on the contrary, 8 alpha, 9 alpha- and 8 beta, 9 beta-EHHCs derived from delta 8-THC may be metabolized by epoxide hydrolase rather than cytochrome P-450 in the human liver, forming 8 beta, 9 alpha-diOH-HHC. -ehhc 45-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 6088753-9 1984 These results indicate that 9 alpha, 10 alpha-EHHC formed from delta 9-THC is further metabolized not by epoxide hydrolase but by monooxygenase system involving cytochrome P-450, and that, on the contrary, 8 alpha, 9 alpha- and 8 beta, 9 beta-EHHCs derived from delta 8-THC may be metabolized by epoxide hydrolase rather than cytochrome P-450 in the human liver, forming 8 beta, 9 alpha-diOH-HHC. -ehhc 45-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 326-342 6088753-9 1984 These results indicate that 9 alpha, 10 alpha-EHHC formed from delta 9-THC is further metabolized not by epoxide hydrolase but by monooxygenase system involving cytochrome P-450, and that, on the contrary, 8 alpha, 9 alpha- and 8 beta, 9 beta-EHHCs derived from delta 8-THC may be metabolized by epoxide hydrolase rather than cytochrome P-450 in the human liver, forming 8 beta, 9 alpha-diOH-HHC. 6,6,9-trimethyl-3-pentyl-6a,7,8,10a-tetrahydrobenzo[c]chromen-1-ol 63-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 6088753-9 1984 These results indicate that 9 alpha, 10 alpha-EHHC formed from delta 9-THC is further metabolized not by epoxide hydrolase but by monooxygenase system involving cytochrome P-450, and that, on the contrary, 8 alpha, 9 alpha- and 8 beta, 9 beta-EHHCs derived from delta 8-THC may be metabolized by epoxide hydrolase rather than cytochrome P-450 in the human liver, forming 8 beta, 9 alpha-diOH-HHC. 6,6,9-trimethyl-3-pentyl-6a,7,8,10a-tetrahydrobenzo[c]chromen-1-ol 63-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 326-342 6422989-0 1984 Selective chemical modification of a functionally linked lysine in cytochrome P-450 LM2. Lysine 57-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 6740183-5 1984 Cytochrome P-450 has a well known sine-qua-non activity in steroid hydroxylations within steroid tissues, and a decrease in the levels of this heme-enzyme might be expected to result in impaired steroidogenesis. Steroids 59-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6740183-5 1984 Cytochrome P-450 has a well known sine-qua-non activity in steroid hydroxylations within steroid tissues, and a decrease in the levels of this heme-enzyme might be expected to result in impaired steroidogenesis. Steroids 89-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6422989-1 1984 Fluorescein isothiocyanate (FITC) has been selectively bound to the epsilon-amino group of lysine-382 in cytochrome P-450 LM2 (RH, reduced-flavoprotein: oxygen oxidoreductase (RH-hydroxylating), EC 1.14.14.1) at pH 8.15. Fluorescein-5-isothiocyanate 0-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 6422989-1 1984 Fluorescein isothiocyanate (FITC) has been selectively bound to the epsilon-amino group of lysine-382 in cytochrome P-450 LM2 (RH, reduced-flavoprotein: oxygen oxidoreductase (RH-hydroxylating), EC 1.14.14.1) at pH 8.15. Fluorescein-5-isothiocyanate 28-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 6422989-2 1984 Benzphetamine N-demethylase activity of the reconstituted FITC-modified cytochrome P-450 LM2 was inhibited by 25%. Fluorescein-5-isothiocyanate 58-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6322709-8 1984 The relative effects of cimetidine and ranitidine on the elimination of cytochrome P-450 metabolized drugs such as theophylline indicate a useful property of ranitidine as compared with cimetidine. Cimetidine 24-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6696254-2 1984 In previous studies they have shown that the halothane-free radical produced by UV-irradiation is identical to that produced during reductive metabolism of halothane by hepatic cytochrome P-450. Halothane 45-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 6696254-2 1984 In previous studies they have shown that the halothane-free radical produced by UV-irradiation is identical to that produced during reductive metabolism of halothane by hepatic cytochrome P-450. Halothane 156-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 6370262-7 1984 When microsomal proteins were separated by sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis, ethanol-fed animals had a distinct band which reflected the increase in microsomal cytochrome P-450 content and seemed to reflect a unique form of cytochrome P-450 induced by ethanol. Ethanol 108-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-207 6370262-7 1984 When microsomal proteins were separated by sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis, ethanol-fed animals had a distinct band which reflected the increase in microsomal cytochrome P-450 content and seemed to reflect a unique form of cytochrome P-450 induced by ethanol. Ethanol 108-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 255-271 6370262-8 1984 Thus, despite the absence of the ADH pathway, a large amount of ethanol was metabolized by MEOS in ADH-negative deermice; this was associated with increased blood ethanol elimination rates, enhanced MEOS activity, and quantitative and qualitative changes of cytochrome P-450. Ethanol 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-274 6697650-2 1984 We used human liver preparations to test whether mephenytoin competes with sparteine for binding to the genetically variable cytochrome P-450, which mediates metabolism of both sparteine and debrisoquine. Mephenytoin 49-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 6697650-2 1984 We used human liver preparations to test whether mephenytoin competes with sparteine for binding to the genetically variable cytochrome P-450, which mediates metabolism of both sparteine and debrisoquine. Sparteine 75-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 6697650-2 1984 We used human liver preparations to test whether mephenytoin competes with sparteine for binding to the genetically variable cytochrome P-450, which mediates metabolism of both sparteine and debrisoquine. Sparteine 177-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 6697650-2 1984 We used human liver preparations to test whether mephenytoin competes with sparteine for binding to the genetically variable cytochrome P-450, which mediates metabolism of both sparteine and debrisoquine. Debrisoquin 191-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 6322709-8 1984 The relative effects of cimetidine and ranitidine on the elimination of cytochrome P-450 metabolized drugs such as theophylline indicate a useful property of ranitidine as compared with cimetidine. Ranitidine 39-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6322709-8 1984 The relative effects of cimetidine and ranitidine on the elimination of cytochrome P-450 metabolized drugs such as theophylline indicate a useful property of ranitidine as compared with cimetidine. Theophylline 115-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6322709-8 1984 The relative effects of cimetidine and ranitidine on the elimination of cytochrome P-450 metabolized drugs such as theophylline indicate a useful property of ranitidine as compared with cimetidine. Ranitidine 158-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6322709-8 1984 The relative effects of cimetidine and ranitidine on the elimination of cytochrome P-450 metabolized drugs such as theophylline indicate a useful property of ranitidine as compared with cimetidine. Cimetidine 186-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6584903-6 1984 For example, phenanthrenequinone was converted to a nonmutagenic metabolite in a cytochrome P-450-dependent reaction, whereas danthron was converted to a highly mutagenic metabolite. 9,10-phenanthrenequinone 13-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 6701906-4 1984 There was a concommitant time-dependent decline in cytochrome P-450 levels in the phenobarbital pretreated animals, but not controls. Phenobarbital 82-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 6422948-7 1984 Cytochrome P-450 destruction in vivo was also greatest in the male rat given norethindrone, whereas no loss was detected in the hen. Norethindrone 77-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6420437-1 1984 6 beta-Hydroxycortisol (6 beta OHF) is a highly polar metabolite of cortisol, probably formed in the endoplasmic reticulum of hepatocytes by cytochrome P-450-dependent microsomal monoxygenases. beta-hydroxycortisol 2-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-157 6696448-1 1984 Complex formation between the phenobarbital-inducible form of rabbit liver microsomal cytochrome P-450 incorporated into phosphatidylcholine and detergent-solubilized cytochrome b5 is associated with a low-to-high spin transition of the former pigment. Phenobarbital 30-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6696448-1 1984 Complex formation between the phenobarbital-inducible form of rabbit liver microsomal cytochrome P-450 incorporated into phosphatidylcholine and detergent-solubilized cytochrome b5 is associated with a low-to-high spin transition of the former pigment. Phosphatidylcholines 121-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6696448-4 1984 Such a cytochrome b5-induced structural alteration of the reconstituted enzyme system is accompanied by an increase in affinity of 4-chloroaniline for cytochrome P-450, as measured in terms of cumene hydroperoxide-supported N-oxidation of the arylamine; the maximum velocity of the catalytic process remains unchanged. 4-chloroaniline 131-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 6696448-4 1984 Such a cytochrome b5-induced structural alteration of the reconstituted enzyme system is accompanied by an increase in affinity of 4-chloroaniline for cytochrome P-450, as measured in terms of cumene hydroperoxide-supported N-oxidation of the arylamine; the maximum velocity of the catalytic process remains unchanged. cumene hydroperoxide 193-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 6696448-4 1984 Such a cytochrome b5-induced structural alteration of the reconstituted enzyme system is accompanied by an increase in affinity of 4-chloroaniline for cytochrome P-450, as measured in terms of cumene hydroperoxide-supported N-oxidation of the arylamine; the maximum velocity of the catalytic process remains unchanged. aniline 243-252 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 6696448-6 1984 Stopped-flow spectrophotometric studies on the influence of cytochrome b5 on the kinetics of binding to cytochrome P-450 of 4-chloroaniline and/or cumene hydroperoxide show that the rates of formation and decay of the adducts change as the molar ratio of cytochrome b5 to cytochrome P-450 varies. 4-chloroaniline 124-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 6696448-6 1984 Stopped-flow spectrophotometric studies on the influence of cytochrome b5 on the kinetics of binding to cytochrome P-450 of 4-chloroaniline and/or cumene hydroperoxide show that the rates of formation and decay of the adducts change as the molar ratio of cytochrome b5 to cytochrome P-450 varies. 4-chloroaniline 124-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 272-288 6420437-1 1984 6 beta-Hydroxycortisol (6 beta OHF) is a highly polar metabolite of cortisol, probably formed in the endoplasmic reticulum of hepatocytes by cytochrome P-450-dependent microsomal monoxygenases. beta ohf 26-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-157 6420437-1 1984 6 beta-Hydroxycortisol (6 beta OHF) is a highly polar metabolite of cortisol, probably formed in the endoplasmic reticulum of hepatocytes by cytochrome P-450-dependent microsomal monoxygenases. Hydrocortisone 14-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-157 6433812-0 1984 Cytochrome P-450-dependent production of chemotactic arachidonate metabolites from human neutrophils. Arachidonic Acid 53-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6708538-0 1984 Effect of human chorionic gonadotropin and estradiol in vivo on estradiol binder and cytochrome P-450 concentrations in rat testis. Estradiol 43-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 6708538-5 1984 Both HCG- and estradiol-induced loss of cytochrome P-450 occur not only in Leydig cells but also in microsomes prepared from seminiferous tubules. Estradiol 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 6595992-3 1984 The loss of cytochrome P-450 cannot be accounted for either by heme destruction or lipid peroxidation under anaerobic conditions. Heme 63-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 6595995-2 1984 These findings exclude the possibility that the inhibitory effect of certain hydroxylation activities of the liver microsomes can be secondary to a direct effect of acrylonitrile on cytochrome P-450 but suggest the hypothesis that the inhibition is mediated by the cyanide released during acrylonitrile metabolism. Acrylonitrile 165-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-198 6525185-0 1984 Effect of the gel-liquid crystalline phase transition on the reduction of cytochrome P-450 reconstituted into dimyristoyl-phosphatidylcholine liposomes. Dimyristoylphosphatidylcholine 110-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 6691964-0 1984 Lateral diffusion of cytochrome P-450 in phospholipid bilayers. Phospholipids 41-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 6146340-7 1984 It is concluded that cimetidine interactions do occur and can be predicted for substances metabolised by the cytochrome P-450 pathway. Cimetidine 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 6331425-1 1984 The rotational motion of the spin labeled substrate analogue n- propylisocyanide bound to the active center of cytochrome P-450 was studied by saturation transfer EPR. n- propylisocyanide 61-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 6091677-0 1984 [Selective chemical modification of cholesterol hydroxylating cytochrome P-450 from adrenal cortex mitochondria by tetranitromethane]. Cholesterol 36-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 6091677-0 1984 [Selective chemical modification of cholesterol hydroxylating cytochrome P-450 from adrenal cortex mitochondria by tetranitromethane]. Tetranitromethane 115-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 6091677-1 1984 Selective chemical modification of adrenocortical cytochrome P-450 responsible for the key stages in steroid biogenesis has been carried out. Steroids 101-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 6378529-3 1984 Recent studies with trace metals have revealed the potent ability of these elements to alter the synthesis and to enhance the degradation of heme moiety of cytochrome P-450. Heme 141-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 6091677-3 1984 Analysis of the cytochrome P-450 nitration kinetics revealed several types of tyrosine residues differing in their accessifility for the reagent. Tyrosine 78-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 6091677-6 1984 Cholesterol and adrenodoxin protected cytochrome P-450 against inactivation by tetranitromethane. Cholesterol 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 6091677-6 1984 Cholesterol and adrenodoxin protected cytochrome P-450 against inactivation by tetranitromethane. Tetranitromethane 79-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 6091677-7 1984 The data obtained are discussed in terms of functional role of tyrosine residues in the cytochrome P-450 molecule. Tyrosine 63-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 6525977-1 1984 The kinetics of cytochrome P-450 content in endoplasmic reticulum membranes at different stages of hepatocarcinogenesis was studied under the influence of unsaturated fatty acids by the EPR method. Fatty Acids, Unsaturated 155-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 6525977-2 1984 At early stages of carcinogenesis the preparations containing oleic, linoleic and arachidonic acids prevent changes in the cytochrome P-450 content usually observed in case of nitrosodiethylamine introduction. oleic 62-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 6525977-2 1984 At early stages of carcinogenesis the preparations containing oleic, linoleic and arachidonic acids prevent changes in the cytochrome P-450 content usually observed in case of nitrosodiethylamine introduction. Linoleic Acid 69-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 6525977-2 1984 At early stages of carcinogenesis the preparations containing oleic, linoleic and arachidonic acids prevent changes in the cytochrome P-450 content usually observed in case of nitrosodiethylamine introduction. Arachidonic Acids 82-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 6525977-2 1984 At early stages of carcinogenesis the preparations containing oleic, linoleic and arachidonic acids prevent changes in the cytochrome P-450 content usually observed in case of nitrosodiethylamine introduction. Diethylnitrosamine 176-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 6525977-3 1984 The diet containing unsaturated fatty acids has a stabilizing effect revealed by the similar changes in the content of cytochrome P-450, phosphatidyl choline and fatty acids composition and also in the level of organization of the endoplasmic reticulum membrane lipid bilayer. Fatty Acids, Unsaturated 20-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 6525977-3 1984 The diet containing unsaturated fatty acids has a stabilizing effect revealed by the similar changes in the content of cytochrome P-450, phosphatidyl choline and fatty acids composition and also in the level of organization of the endoplasmic reticulum membrane lipid bilayer. Fatty Acids 32-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 6326394-0 1984 The influence of oxygen donor ligation on the spectroscopic properties of ferric cytochrome P-450: ester, ether and ketone co-ordination to the haem iron. Oxygen 17-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 6533032-0 1984 Metabolism of nitrosamines by cytochrome P-450 isozymes. Nitrosamines 14-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 6326394-0 1984 The influence of oxygen donor ligation on the spectroscopic properties of ferric cytochrome P-450: ester, ether and ketone co-ordination to the haem iron. Esters 99-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 6426177-0 1984 Rate-limiting steps in cytochrome P-450-catalysed reactions: studies on isotope effects in the O-de-ethylation of 7-ethoxycoumarin. 7-ethoxycoumarin 114-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 6326394-0 1984 The influence of oxygen donor ligation on the spectroscopic properties of ferric cytochrome P-450: ester, ether and ketone co-ordination to the haem iron. Ether 106-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 6426177-1 1984 Primary deuterium isotope effects ranging from 2 to 6 were measured for the O-de-ethylation of 7-ethoxycoumarin catalysed by microsomal and purified cytochrome P-450 isozymes. Deuterium 8-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 6326394-0 1984 The influence of oxygen donor ligation on the spectroscopic properties of ferric cytochrome P-450: ester, ether and ketone co-ordination to the haem iron. Ketones 116-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 6426177-1 1984 Primary deuterium isotope effects ranging from 2 to 6 were measured for the O-de-ethylation of 7-ethoxycoumarin catalysed by microsomal and purified cytochrome P-450 isozymes. 7-ethoxycoumarin 95-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 6326394-0 1984 The influence of oxygen donor ligation on the spectroscopic properties of ferric cytochrome P-450: ester, ether and ketone co-ordination to the haem iron. Iron 149-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 6326394-1 1984 Homogeneous low-spin complexes of cytochrome P-450-CAM with esters, ethers and ketones have been prepared and characterized by u.v.-visible absorption, circular dichroism (CD), magnetic circular dichroism (MCD) and electron paramagnetic resonance (EPR) spectroscopy. Esters 60-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 6326394-1 1984 Homogeneous low-spin complexes of cytochrome P-450-CAM with esters, ethers and ketones have been prepared and characterized by u.v.-visible absorption, circular dichroism (CD), magnetic circular dichroism (MCD) and electron paramagnetic resonance (EPR) spectroscopy. Ethers 68-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 6326394-1 1984 Homogeneous low-spin complexes of cytochrome P-450-CAM with esters, ethers and ketones have been prepared and characterized by u.v.-visible absorption, circular dichroism (CD), magnetic circular dichroism (MCD) and electron paramagnetic resonance (EPR) spectroscopy. Ketones 79-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 6326394-8 1984 Anomalous spectral and substrate binding properties have been reported in the study of cytochrome P-450 under conditions employing solvents and non-phosphate buffers containing oxygen functionalities, and have been attributed to "solvent effects". Phosphates 148-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-103 6326394-8 1984 Anomalous spectral and substrate binding properties have been reported in the study of cytochrome P-450 under conditions employing solvents and non-phosphate buffers containing oxygen functionalities, and have been attributed to "solvent effects". Oxygen 177-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-103 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Alcohols 61-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Alcohols 61-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-259 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Amides 70-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Amides 70-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-259 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. carboxylate 80-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. carboxylate 80-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-259 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Oxygen 92-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Oxygen 92-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-259 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Oxygen 171-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Oxygen 171-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-259 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Iron 235-239 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 6326394-9 1984 The present work, in combination with our previous report of alcohol, amide and carboxylate oxygen donor complexes of cytochrome P-450, is evidence that a wide variety of oxygen-donor species are capable of direct ligation to the haem iron of cytochrome P-450. Iron 235-239 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-259 6326394-10 1984 This leads us to suggest oxygen-donor ligation to cytochrome P-450 as the origin of spectral and substrate binding anomalies previously attributed to solvent effects. Oxygen 25-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 6372265-7 1984 The polymorphism in the 4-hydroxylation of debrisoquine observed in vivo has been shown to be due to a defect in a specific form of cytochrome P-450 which appears to be under monogenic regulation. Debrisoquin 43-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 6661247-0 1983 Hepatic microsomal cytochrome p-450-dependent N-demethylation of methylguanidine. Nitrogen 46-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 6372266-0 1984 Cytochrome P-450 spin state: inorganic biochemistry of haem iron ligation and functional significance. Iron 60-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6661247-0 1983 Hepatic microsomal cytochrome p-450-dependent N-demethylation of methylguanidine. Methylguanidine 65-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 6372267-1 1984 This review presents current ideas, models and experimental data relating to the precise chemistry that links the transition metal active centre of cytochrome P-450 systems, the unactivated alkane substrate and the triplet atmospheric dioxygen molecule. Metals 125-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 6661247-1 1983 Cytochrome P-450-dependent N-demethylation of methylguanidine, a uremia toxin, was investigated. Methylguanidine 46-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6372267-1 1984 This review presents current ideas, models and experimental data relating to the precise chemistry that links the transition metal active centre of cytochrome P-450 systems, the unactivated alkane substrate and the triplet atmospheric dioxygen molecule. Alkanes 190-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Nitrogen 50-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Nitrogen 50-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 6372267-1 1984 This review presents current ideas, models and experimental data relating to the precise chemistry that links the transition metal active centre of cytochrome P-450 systems, the unactivated alkane substrate and the triplet atmospheric dioxygen molecule. Oxygen 235-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Nitrogen 50-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 6661254-1 1983 Bis(tri-n-butyltin)oxide, an agriculturally important biocidal agent, when added in vitro to liver microsomes containing the phenobarbital-induced form of cytochrome P-450, produced a typical type I binding spectrum (an absorption maximum at 390 nm; an absorption minimum at 420 nm). bis(tri-n-butyltin)oxide 0-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Methylguanidine 116-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Methylguanidine 116-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Methylguanidine 116-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Carbon Monoxide 254-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Carbon Monoxide 254-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 6661254-1 1983 Bis(tri-n-butyltin)oxide, an agriculturally important biocidal agent, when added in vitro to liver microsomes containing the phenobarbital-induced form of cytochrome P-450, produced a typical type I binding spectrum (an absorption maximum at 390 nm; an absorption minimum at 420 nm). Phenobarbital 125-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Carbon Monoxide 254-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Metyrapone 274-284 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 6661254-2 1983 Studies with microsomal preparations containing cytochrome P-448, induced by 3-methylcholanthrene or beta-naphthoflavone, revealed that this hemeprotein was more susceptible to direct degradation by bis(tri-n-butyltin)oxide than was the uninduced or phenobarbital-induced forms of cytochrome P-450. bis(tri-n-butyltin)oxide 199-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 281-297 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Metyrapone 274-284 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 6661247-5 1983 The direct involvement of cytochrome P-450 in the N-demethylation is supported by the observations that addition of methylguanidine to purified cytochrome P-450 preparation caused a type I spectral change and that inhibitors of cytochrome P-450, such as carbon monoxide and metyrapone, markedly decreased the rate of demethylation. Metyrapone 274-284 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 6661247-7 1983 In addition, guanidine formation was observed in the reconstituted system containing purified cytochrome P-450. Guanidine 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 6661254-4 1983 The formation of cytochrome P-420 was both time and temperature dependent, and it also occurred to a greater extent in microsomal preparations containing cytochrome P-448 than in microsomes containing the phenobarbital-induced form of cytochrome P-450. Phenobarbital 205-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 235-251 6584878-0 1983 Nutrition-endocrine interactions: induction of reciprocal changes in the delta 4-5 alpha-reduction of testosterone and the cytochrome P-450-dependent oxidation of estradiol by dietary macronutrients in man. Estradiol 163-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 6196179-5 1983 In correlation with the changes in these enzyme activities induced by hCG, the concentration of ovarian microsomal cytochrome P-450 decreased to barely detectable level after hCG treatment; this decrease was also prevented by pretreatment with cycloheximide. Cycloheximide 244-257 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 6584878-6 1983 The mechanism by which reciprocal changes in the delta 4-5 alpha-reduction of testosterone and the cytochrome P-450-mediated oxidation of estradiol are produced by diets is not known. Estradiol 138-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 6584878-4 1983 The delta 4-5 alpha-reduction of testosterone was considerably diminished, while the cytochrome P-450-dependent hydroxylation of estradiol at the C2 position was substantially enhanced during ingestion of the high-protein diet as compared with the high-carbohydrate diet. Estradiol 129-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 6584878-7 1983 Similar changes in steroid delta 4-5 alpha-reduction and cytochrome P-450-dependent chemical oxidations have been observed in circumstances in which the mixed-function oxidase system in liver is induced by agents such as phenobarbital, hexachlorobenzene, dioxin, and polyhalogenated biphenyls. Phenobarbital 221-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 6584878-4 1983 The delta 4-5 alpha-reduction of testosterone was considerably diminished, while the cytochrome P-450-dependent hydroxylation of estradiol at the C2 position was substantially enhanced during ingestion of the high-protein diet as compared with the high-carbohydrate diet. Carbohydrates 253-265 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 6584878-7 1983 Similar changes in steroid delta 4-5 alpha-reduction and cytochrome P-450-dependent chemical oxidations have been observed in circumstances in which the mixed-function oxidase system in liver is induced by agents such as phenobarbital, hexachlorobenzene, dioxin, and polyhalogenated biphenyls. Hexachlorobenzene 236-253 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 6639949-6 1983 Comparison to ferric myoglobin-thiolate complexes indicates that the negative sign observed for the cytochrome P-450 spectra is not a property of the thiolate fifth ligand, but rather arises from a different interaction of the cytochrome P-450 heme with its protein environment. thiolate 31-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 6584878-7 1983 Similar changes in steroid delta 4-5 alpha-reduction and cytochrome P-450-dependent chemical oxidations have been observed in circumstances in which the mixed-function oxidase system in liver is induced by agents such as phenobarbital, hexachlorobenzene, dioxin, and polyhalogenated biphenyls. Dioxins 255-261 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 6584878-7 1983 Similar changes in steroid delta 4-5 alpha-reduction and cytochrome P-450-dependent chemical oxidations have been observed in circumstances in which the mixed-function oxidase system in liver is induced by agents such as phenobarbital, hexachlorobenzene, dioxin, and polyhalogenated biphenyls. polyhalogenated biphenyls 267-292 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 6639949-6 1983 Comparison to ferric myoglobin-thiolate complexes indicates that the negative sign observed for the cytochrome P-450 spectra is not a property of the thiolate fifth ligand, but rather arises from a different interaction of the cytochrome P-450 heme with its protein environment. Heme 244-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 6639949-6 1983 Comparison to ferric myoglobin-thiolate complexes indicates that the negative sign observed for the cytochrome P-450 spectra is not a property of the thiolate fifth ligand, but rather arises from a different interaction of the cytochrome P-450 heme with its protein environment. thiolate 31-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 227-243 6639949-6 1983 Comparison to ferric myoglobin-thiolate complexes indicates that the negative sign observed for the cytochrome P-450 spectra is not a property of the thiolate fifth ligand, but rather arises from a different interaction of the cytochrome P-450 heme with its protein environment. Heme 244-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 227-243 6639949-6 1983 Comparison to ferric myoglobin-thiolate complexes indicates that the negative sign observed for the cytochrome P-450 spectra is not a property of the thiolate fifth ligand, but rather arises from a different interaction of the cytochrome P-450 heme with its protein environment. thiolate 150-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 6661451-0 1983 [Comparative inhibitory analysis of aniline hydroxylation by cytochrome P-450 in NADPH-, hydroperoxide cumyl- and H2O2-dependent systems]. aniline 36-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6639949-8 1983 Hyperporphyrin (split Soret) cytochrome P-450 complexes with thiolates, phosphines and cyanide trans to cysteinate have complex CD spectra, reflecting the intrinsic non-degeneracy of the Soret pi pi transitions. hyperporphyrin 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 6639949-8 1983 Hyperporphyrin (split Soret) cytochrome P-450 complexes with thiolates, phosphines and cyanide trans to cysteinate have complex CD spectra, reflecting the intrinsic non-degeneracy of the Soret pi pi transitions. thiolates 61-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 6639949-8 1983 Hyperporphyrin (split Soret) cytochrome P-450 complexes with thiolates, phosphines and cyanide trans to cysteinate have complex CD spectra, reflecting the intrinsic non-degeneracy of the Soret pi pi transitions. Phosphines 72-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 6639949-8 1983 Hyperporphyrin (split Soret) cytochrome P-450 complexes with thiolates, phosphines and cyanide trans to cysteinate have complex CD spectra, reflecting the intrinsic non-degeneracy of the Soret pi pi transitions. Cyanides 87-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 6639949-8 1983 Hyperporphyrin (split Soret) cytochrome P-450 complexes with thiolates, phosphines and cyanide trans to cysteinate have complex CD spectra, reflecting the intrinsic non-degeneracy of the Soret pi pi transitions. cysteinate 104-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 6640098-2 1983 LPO activation in hepatocyte suspension by a catalytic system containing Fe2+--ADP plus NADP X H makes the destruction of cytochrome P-450 more rapid. fe2+--adp 73-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 6640098-2 1983 LPO activation in hepatocyte suspension by a catalytic system containing Fe2+--ADP plus NADP X H makes the destruction of cytochrome P-450 more rapid. nadp x h 88-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 6661451-0 1983 [Comparative inhibitory analysis of aniline hydroxylation by cytochrome P-450 in NADPH-, hydroperoxide cumyl- and H2O2-dependent systems]. NADP 81-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6640098-3 1983 Supplementation of the incubation medium with the antioxidant, 2-ethyl-6-methyl-3-hydroxypyridine (HP-6), inhibits LPO, on the one hand, and stabilizes cytochrome P-450, on the other one. 6-methyl-2-ethyl-3-hydroxypyridine 63-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-168 6661451-0 1983 [Comparative inhibitory analysis of aniline hydroxylation by cytochrome P-450 in NADPH-, hydroperoxide cumyl- and H2O2-dependent systems]. Hydrogen Peroxide 89-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6640098-3 1983 Supplementation of the incubation medium with the antioxidant, 2-ethyl-6-methyl-3-hydroxypyridine (HP-6), inhibits LPO, on the one hand, and stabilizes cytochrome P-450, on the other one. hp-6 99-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-168 6661451-0 1983 [Comparative inhibitory analysis of aniline hydroxylation by cytochrome P-450 in NADPH-, hydroperoxide cumyl- and H2O2-dependent systems]. Hydrogen Peroxide 114-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6661451-1 1983 The p-hydroxylation of aniline in the presence of liver microsomes and isolated cytochrome P-450 in NADPH-, H2O2- and cumyl hydroperoxide (CHP)-dependent systems was investigated. NADP 100-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 6661451-1 1983 The p-hydroxylation of aniline in the presence of liver microsomes and isolated cytochrome P-450 in NADPH-, H2O2- and cumyl hydroperoxide (CHP)-dependent systems was investigated. Hydrogen Peroxide 108-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 6661451-1 1983 The p-hydroxylation of aniline in the presence of liver microsomes and isolated cytochrome P-450 in NADPH-, H2O2- and cumyl hydroperoxide (CHP)-dependent systems was investigated. cumene hydroperoxide 118-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 6661451-1 1983 The p-hydroxylation of aniline in the presence of liver microsomes and isolated cytochrome P-450 in NADPH-, H2O2- and cumyl hydroperoxide (CHP)-dependent systems was investigated. cumene hydroperoxide 139-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 6661451-4 1983 Since the kcat value for the H2O2-dependent system is much higher than that for the NADPH- or CHP-systems, it may be assumed that when H2O2 is used as a cosubstrate for this reaction, the true value of the degradation constant for the cytochrome P-450--O2(2-)--aniline peroxycomplex can be measured. Hydrogen Peroxide 29-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 235-251 6661451-4 1983 Since the kcat value for the H2O2-dependent system is much higher than that for the NADPH- or CHP-systems, it may be assumed that when H2O2 is used as a cosubstrate for this reaction, the true value of the degradation constant for the cytochrome P-450--O2(2-)--aniline peroxycomplex can be measured. NADP 84-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 235-251 6661451-4 1983 Since the kcat value for the H2O2-dependent system is much higher than that for the NADPH- or CHP-systems, it may be assumed that when H2O2 is used as a cosubstrate for this reaction, the true value of the degradation constant for the cytochrome P-450--O2(2-)--aniline peroxycomplex can be measured. Hydrogen Peroxide 135-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 235-251 6661451-4 1983 Since the kcat value for the H2O2-dependent system is much higher than that for the NADPH- or CHP-systems, it may be assumed that when H2O2 is used as a cosubstrate for this reaction, the true value of the degradation constant for the cytochrome P-450--O2(2-)--aniline peroxycomplex can be measured. o2(2-)--aniline 253-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 235-251 6684689-1 1983 Previous studies have shown that 6-thiopurine is metabolically activated by hepatic cytochrome P-450 to an intermediate capable of binding to proteins by a mixed disulfide linkage. Mercaptopurine 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 6357293-3 1983 The interaction of this protein and of the native protein with cholesterol-specific cytochrome P-450 and adrenodoxin reductase occurs in a similar way. Cholesterol 63-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 6684689-1 1983 Previous studies have shown that 6-thiopurine is metabolically activated by hepatic cytochrome P-450 to an intermediate capable of binding to proteins by a mixed disulfide linkage. Disulfides 162-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 6626210-1 1983 The filamentous fungus Aspergillus ochraceus TS produces an inducible microsomal cytochrome P-450 linked monooxygenase which is capable of hydroxylating benzo(a)pyrene in presence of O2 and NADPH. Benzo(a)pyrene 153-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 6888166-0 1983 Inhibition of lipid peroxide decomposition by compounds which bind with cytochrome p-450. Lipid Peroxides 14-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6888166-2 1983 The results indicate that the in vitro interaction of hexobarbital and SKF-525 A (type I binding compounds) with microsomal cytochrome p-450 inhibits the peroxidase activity while the in vitro interaction of aniline (type II binding compound) only slightly affect the peroxidase activity. Hexobarbital 54-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 6888166-2 1983 The results indicate that the in vitro interaction of hexobarbital and SKF-525 A (type I binding compounds) with microsomal cytochrome p-450 inhibits the peroxidase activity while the in vitro interaction of aniline (type II binding compound) only slightly affect the peroxidase activity. aniline 208-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 6626210-1 1983 The filamentous fungus Aspergillus ochraceus TS produces an inducible microsomal cytochrome P-450 linked monooxygenase which is capable of hydroxylating benzo(a)pyrene in presence of O2 and NADPH. Oxygen 183-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 6626210-1 1983 The filamentous fungus Aspergillus ochraceus TS produces an inducible microsomal cytochrome P-450 linked monooxygenase which is capable of hydroxylating benzo(a)pyrene in presence of O2 and NADPH. NADP 190-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 6626210-5 1983 These findings together with the results obtained with flavone on the metabolism of benzo(a)pyrene by various microsomal preparations suggest a parallel induction of multiple forms of cytochrome P-450 as observed in mammalian liver under identical condition. flavone 55-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-200 6626210-5 1983 These findings together with the results obtained with flavone on the metabolism of benzo(a)pyrene by various microsomal preparations suggest a parallel induction of multiple forms of cytochrome P-450 as observed in mammalian liver under identical condition. Benzo(a)pyrene 84-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-200 6409396-1 1983 There are two major R,S-1-(tetrahydro-2-furanyl)-5-fluorouracil (ftorafur) activation pathways to 5-fluorouracil, one that is mediated by microsomal cytochrome P-450 oxidation at C-5" of the tetrahydrofuran moiety and one that is mediated by soluble enzymes. r,s-1-(tetrahydro-2-furanyl)-5-fluorouracil 20-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 6409396-1 1983 There are two major R,S-1-(tetrahydro-2-furanyl)-5-fluorouracil (ftorafur) activation pathways to 5-fluorouracil, one that is mediated by microsomal cytochrome P-450 oxidation at C-5" of the tetrahydrofuran moiety and one that is mediated by soluble enzymes. Fluorouracil 49-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 6409396-1 1983 There are two major R,S-1-(tetrahydro-2-furanyl)-5-fluorouracil (ftorafur) activation pathways to 5-fluorouracil, one that is mediated by microsomal cytochrome P-450 oxidation at C-5" of the tetrahydrofuran moiety and one that is mediated by soluble enzymes. Tegafur 65-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 6409396-1 1983 There are two major R,S-1-(tetrahydro-2-furanyl)-5-fluorouracil (ftorafur) activation pathways to 5-fluorouracil, one that is mediated by microsomal cytochrome P-450 oxidation at C-5" of the tetrahydrofuran moiety and one that is mediated by soluble enzymes. tetrahydrofuran 191-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 6871822-4 1983 Dimethylhydrazine metabolism in each segment is inhibited significantly by inhibitors of the cytochrome P-450-dependent mixed function oxidase system. Dimethylhydrazines 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 6411467-3 1983 The cytochrome P-450 content and response to induction by 3-methylcholanthrene and phenobarbitone; the distribution of lactate dehydrogenase, glucose-6-phosphatase, pyruvate kinase and tyrosine aminotransferase activities in the sub-populations is also reported. Methylcholanthrene 58-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 6411467-3 1983 The cytochrome P-450 content and response to induction by 3-methylcholanthrene and phenobarbitone; the distribution of lactate dehydrogenase, glucose-6-phosphatase, pyruvate kinase and tyrosine aminotransferase activities in the sub-populations is also reported. Phenobarbital 83-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 6411097-0 1983 Formation of electrophilic chlorine from carbon tetrachloride--involvement of cytochrome P-450. Chlorine 27-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 6411097-0 1983 Formation of electrophilic chlorine from carbon tetrachloride--involvement of cytochrome P-450. Carbon Tetrachloride 41-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 6136493-6 1983 Endotoxin administration was also shown to diminish, but not prevent, the induction of cytochrome P--450 and ethylmorphine N-demethylase activity produced by phenobarbital. Phenobarbital 158-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-104 6412745-0 1983 Possible association of NADPH-cytochrome P-450 reductase and cytochrome P-450 in reconstituted phospholipid vesicles. Phospholipids 95-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 6407834-0 1983 Reduction of cytochrome P-450 LM2 by NADPH in reconstituted phospholipid vesicles is dependent on membrane charge. NADP 37-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 6407834-0 1983 Reduction of cytochrome P-450 LM2 by NADPH in reconstituted phospholipid vesicles is dependent on membrane charge. Phospholipids 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 6407834-1 1983 The kinetics of the reduction of cytochrome P-450 LM2 mediated by NADPH-cytochrome P-450 reductase in reconstituted phospholipid vesicles was examined. Phospholipids 116-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 6409115-5 1983 These data suggest that the discordant effects of phenobarbital on UDPGT and cytochrome P-450 previously reported in humans and rats may not be attributable solely to differences in the drug doses employed. Phenobarbital 50-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 6407834-4 1983 In the presence of benzphetamine, the extent of cytochrome P-450 LM2 reduced 1 s after the addition of NADPH to the system was a linear function of the electrophoretic mobilities of the vesicles used. Benzphetamine 19-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 6407834-4 1983 In the presence of benzphetamine, the extent of cytochrome P-450 LM2 reduced 1 s after the addition of NADPH to the system was a linear function of the electrophoretic mobilities of the vesicles used. NADP 103-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 6872096-5 1983 BHA addition effectively discharged the activated oxygen complex of cytochrome P-450 (liver microsomes) as well as Comp. bha 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 6305968-5 1983 Transfer of cholesterol to and from cytochrome P-450 occurred with similar first order rate constants and was also independent of the concentrations of cholesterol vesicles and P-450. Cholesterol 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 6872096-5 1983 BHA addition effectively discharged the activated oxygen complex of cytochrome P-450 (liver microsomes) as well as Comp. Oxygen 50-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 6354819-6 1983 NADPH is also the main donor for reducing equivalents in drug oxidations by the cytochrome P-450-dependent monooxygenase system which, with some notable exceptions, serves important purposes in detoxication. NADP 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 6309256-1 1983 The iodine-containing stable iminoxyl radicals with various distances between the N-O-group and the iodine atom are proposed to be used to study the structure of the active center of the microsomal cytochrome P-450. Iodine 4-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-214 6859878-1 1983 Epoxyeicosatrienoic acids, formed during the cytochrome P-450-catalyzed oxidation of arachidonic acid, react with a liver cytosolic epoxide hydrolase to form vicinal diols of eicosatrienoic acid. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 6309256-1 1983 The iodine-containing stable iminoxyl radicals with various distances between the N-O-group and the iodine atom are proposed to be used to study the structure of the active center of the microsomal cytochrome P-450. iminoxyl 29-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-214 6859878-1 1983 Epoxyeicosatrienoic acids, formed during the cytochrome P-450-catalyzed oxidation of arachidonic acid, react with a liver cytosolic epoxide hydrolase to form vicinal diols of eicosatrienoic acid. Arachidonic Acid 85-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 6859878-1 1983 Epoxyeicosatrienoic acids, formed during the cytochrome P-450-catalyzed oxidation of arachidonic acid, react with a liver cytosolic epoxide hydrolase to form vicinal diols of eicosatrienoic acid. vicinal diols 158-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 6309256-1 1983 The iodine-containing stable iminoxyl radicals with various distances between the N-O-group and the iodine atom are proposed to be used to study the structure of the active center of the microsomal cytochrome P-450. Iodine 100-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-214 6859878-1 1983 Epoxyeicosatrienoic acids, formed during the cytochrome P-450-catalyzed oxidation of arachidonic acid, react with a liver cytosolic epoxide hydrolase to form vicinal diols of eicosatrienoic acid. Eicosatrienoic acid 5-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 6309256-2 1983 The radicals used induce changes in the optical spectra of the Fe3+ ion located in the active center of the enzyme, as in the case of type 1 substrates and inhibit essentially the microsomal oxidation of cytochrome P-450 substrates of type 1 and 2. ferric sulfate 63-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-220 6309256-4 1983 Cytochrome P-450 substrates (aminopyrine) protect the enzyme against the radical-induced inactivation. Aminopyrine 29-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6309256-4 1983 Cytochrome P-450 substrates (aminopyrine) protect the enzyme against the radical-induced inactivation. radical 73-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6309256-5 1983 The iodine-containing radicals are covalently bound to cytochrome P-450 in the vicinity of active center. Iodine 4-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 6309256-7 1983 The EPR method was used to detect the coupling between Fe3+ and the radical located in the active center of cytochrome P-450. ferric sulfate 55-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 6309256-8 1983 The saturation curves of radical SPR spectra at 77 degrees K were employed to determine the contribution of Fe3+ to the relaxation time, T1, of the radicals covalently bound to cytochrome P-450 and to estimate the distances between the Fe3+ ion and the N-O-group of these radicals in the enzyme active center. ferric sulfate 108-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 6309256-8 1983 The saturation curves of radical SPR spectra at 77 degrees K were employed to determine the contribution of Fe3+ to the relaxation time, T1, of the radicals covalently bound to cytochrome P-450 and to estimate the distances between the Fe3+ ion and the N-O-group of these radicals in the enzyme active center. ferric sulfate 236-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 6870907-0 1983 Correlations between spin equilibrium shift, reduction rate, and N-demethylation activity in liver microsomal cytochrome P-450 and a series of benzphetamine analogues as substrates. Nitrogen 65-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 6860533-4 1983 It is proposed that sulphinpyrazone has differential effects on at least two forms of cytochrome P-450 inhibiting one enzyme or group of enzymes which metabolises tolbutamide, phenytoin and S(-)warfarin and inducing a form (or forms) which metabolises theophylline, antipyrine and R(+)warfarin. Tolbutamide 163-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6860533-4 1983 It is proposed that sulphinpyrazone has differential effects on at least two forms of cytochrome P-450 inhibiting one enzyme or group of enzymes which metabolises tolbutamide, phenytoin and S(-)warfarin and inducing a form (or forms) which metabolises theophylline, antipyrine and R(+)warfarin. Phenytoin 176-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6860533-4 1983 It is proposed that sulphinpyrazone has differential effects on at least two forms of cytochrome P-450 inhibiting one enzyme or group of enzymes which metabolises tolbutamide, phenytoin and S(-)warfarin and inducing a form (or forms) which metabolises theophylline, antipyrine and R(+)warfarin. Sulfinpyrazone 20-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6860533-4 1983 It is proposed that sulphinpyrazone has differential effects on at least two forms of cytochrome P-450 inhibiting one enzyme or group of enzymes which metabolises tolbutamide, phenytoin and S(-)warfarin and inducing a form (or forms) which metabolises theophylline, antipyrine and R(+)warfarin. Warfarin 190-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6860533-4 1983 It is proposed that sulphinpyrazone has differential effects on at least two forms of cytochrome P-450 inhibiting one enzyme or group of enzymes which metabolises tolbutamide, phenytoin and S(-)warfarin and inducing a form (or forms) which metabolises theophylline, antipyrine and R(+)warfarin. Theophylline 252-264 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6860533-4 1983 It is proposed that sulphinpyrazone has differential effects on at least two forms of cytochrome P-450 inhibiting one enzyme or group of enzymes which metabolises tolbutamide, phenytoin and S(-)warfarin and inducing a form (or forms) which metabolises theophylline, antipyrine and R(+)warfarin. Antipyrine 266-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6860533-4 1983 It is proposed that sulphinpyrazone has differential effects on at least two forms of cytochrome P-450 inhibiting one enzyme or group of enzymes which metabolises tolbutamide, phenytoin and S(-)warfarin and inducing a form (or forms) which metabolises theophylline, antipyrine and R(+)warfarin. Warfarin 281-293 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6833253-0 1983 Stereochemistry of cytochrome P-450-catalyzed epoxidation and prosthetic heme alkylation. Heme 73-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 6833253-1 1983 Oxidation of 1-octene by cytochrome P-450 results concurrently in formation of 1,2-oxidooctane and in N-alkylation by the catalytically activated olefin of the prosthetic heme group. 1-octene 13-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 6833253-1 1983 Oxidation of 1-octene by cytochrome P-450 results concurrently in formation of 1,2-oxidooctane and in N-alkylation by the catalytically activated olefin of the prosthetic heme group. 1,2-oxidooctane 79-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 6833253-1 1983 Oxidation of 1-octene by cytochrome P-450 results concurrently in formation of 1,2-oxidooctane and in N-alkylation by the catalytically activated olefin of the prosthetic heme group. Alkenes 146-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 6833253-1 1983 Oxidation of 1-octene by cytochrome P-450 results concurrently in formation of 1,2-oxidooctane and in N-alkylation by the catalytically activated olefin of the prosthetic heme group. Heme 171-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 6831832-4 1983 This suggests increasing theophylline metabolism with age due to developing hepatic cytochrome P-450 enzyme systems. Theophylline 25-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 6617864-0 1983 Effect of succinylacetone on heme and cytochrome P450 synthesis in hepatocyte culture. succinylacetone 10-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 6833217-1 1983 We have employed small angle x-ray scattering to examine the solution conformation of cytochrome P-450cam in the presence and absence of its substrate, camphor. Camphor 152-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 6617864-1 1983 The effects of succinylacetone, a tyrosine metabolite, on the hepatic biosynthesis of heme and cytochrome P450 were studied in primary culture of chick embryo hepatocytes. succinylacetone 15-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 6617864-2 1983 Succinylacetone potentiated the phenobarbital-mediated induction of delta-aminolevulinate synthase, strongly inhibited porphobilinogen synthase activity, reduced cellular heme concentration and impaired induction of cytochrome P450. succinylacetone 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-231 6617864-2 1983 Succinylacetone potentiated the phenobarbital-mediated induction of delta-aminolevulinate synthase, strongly inhibited porphobilinogen synthase activity, reduced cellular heme concentration and impaired induction of cytochrome P450. Phenobarbital 32-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-231 6617864-3 1983 Enhanced induction of delta-aminolevulinate synthase and decreased cytochrome P450 induction may be explained by the succinylacetone-mediated inhibition of porphobilinogen synthase and the subsequent depletion of intracellular heme, since these effects of succinylacetone were reversed by addition of heme. succinylacetone 117-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 6617864-3 1983 Enhanced induction of delta-aminolevulinate synthase and decreased cytochrome P450 induction may be explained by the succinylacetone-mediated inhibition of porphobilinogen synthase and the subsequent depletion of intracellular heme, since these effects of succinylacetone were reversed by addition of heme. Heme 227-231 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 6401621-1 1983 FSH, estradiol, or a combination of FSH and estradiol enhanced secretion of progesterone by primary cultures of immature porcine granulosa cells; they also increased the mitochondrial content of cytochrome P-450 and mitochondrial cholesterol side-chain cleavage activity. Estradiol 5-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-211 6838647-4 1983 In contrast, lidocaine and its derivatives (bupivacaine, mepivacaine, etidocaine, pyrrocaine and prilocaine) were found to induce delta-aminolevulinate synthetase and to cause accumulation of porphyrins and cytochrome P-450. Lidocaine 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 6838647-4 1983 In contrast, lidocaine and its derivatives (bupivacaine, mepivacaine, etidocaine, pyrrocaine and prilocaine) were found to induce delta-aminolevulinate synthetase and to cause accumulation of porphyrins and cytochrome P-450. Bupivacaine 44-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 6838647-4 1983 In contrast, lidocaine and its derivatives (bupivacaine, mepivacaine, etidocaine, pyrrocaine and prilocaine) were found to induce delta-aminolevulinate synthetase and to cause accumulation of porphyrins and cytochrome P-450. Mepivacaine 57-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 6838647-4 1983 In contrast, lidocaine and its derivatives (bupivacaine, mepivacaine, etidocaine, pyrrocaine and prilocaine) were found to induce delta-aminolevulinate synthetase and to cause accumulation of porphyrins and cytochrome P-450. Etidocaine 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 6838647-4 1983 In contrast, lidocaine and its derivatives (bupivacaine, mepivacaine, etidocaine, pyrrocaine and prilocaine) were found to induce delta-aminolevulinate synthetase and to cause accumulation of porphyrins and cytochrome P-450. pyrrocaine 82-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 6838647-4 1983 In contrast, lidocaine and its derivatives (bupivacaine, mepivacaine, etidocaine, pyrrocaine and prilocaine) were found to induce delta-aminolevulinate synthetase and to cause accumulation of porphyrins and cytochrome P-450. Prilocaine 97-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 6838653-1 1983 In rats, erythromycin has been shown to induce microsomal enzymes and to promote its own transformation into a metabolite which forms an inactive complex with reduced cytochrome P-450. Erythromycin 9-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-183 6838653-3 1983 In the treated patients, NADPH-cytochrome c reductase activity was increased; the total cytochrome P-450 concn was also increased but part of the total cytochrome P-450 was complexed by an erythromycin metabolite. Erythromycin 189-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-168 6838653-6 1983 We conclude that the administration of erythromycin propionate induces microsomal enzymes and results in the formation of an inactive cytochrome P-450-metabolite complex in humans. erythromycin propionate 39-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 6401621-1 1983 FSH, estradiol, or a combination of FSH and estradiol enhanced secretion of progesterone by primary cultures of immature porcine granulosa cells; they also increased the mitochondrial content of cytochrome P-450 and mitochondrial cholesterol side-chain cleavage activity. Estradiol 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-211 6185486-9 1983 The decay of activated bleomycin yields low spin ferric bleomycin, a complex with Mossbauer parameters nearly identical with those reported for ferric cytochrome P-450. iron bleomycin 49-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 6826727-7 1983 In cultured hepatocytes, succinylacetone also inhibited ALA dehydratase activity, decreased the cellular content of heme and cytochrome P-450, and greatly potentiated the induction response of ALA synthase to drugs such as phenobarbital, chemicals such as allylisopropylacetamide and 3,5-dicarbethoxy-1,4-dihydrocollidine, and natural steroids such as etiocholanolone. succinylacetone 25-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 6220203-0 1983 Substrate specificity of the form of cytochrome P-450 catalyzing the 4-hydroxylation of debrisoquine in man. Debrisoquin 88-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 6220203-1 1983 In the present study we have investigated the substrate specificity of the form of cytochrome P-450 catalyzing the 4-hydroxylation of debrisoquine in man by analyzing the kinetics of inhibition of this activity by potential alternative substrates for the enzyme. Debrisoquin 134-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 6185486-10 1983 Although iron bleomycin does not have a polyaromatic structure like heme, many features of its electronic structure at the iron are very similar to those produced by the sulfur-coordinated heme iron of ferric cytochrome P-450, a protein that catalyzes a similar oxygen-dependent reaction. iron bleomycin 9-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 6185486-10 1983 Although iron bleomycin does not have a polyaromatic structure like heme, many features of its electronic structure at the iron are very similar to those produced by the sulfur-coordinated heme iron of ferric cytochrome P-450, a protein that catalyzes a similar oxygen-dependent reaction. Heme 68-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 6185486-10 1983 Although iron bleomycin does not have a polyaromatic structure like heme, many features of its electronic structure at the iron are very similar to those produced by the sulfur-coordinated heme iron of ferric cytochrome P-450, a protein that catalyzes a similar oxygen-dependent reaction. Iron 9-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 6185486-10 1983 Although iron bleomycin does not have a polyaromatic structure like heme, many features of its electronic structure at the iron are very similar to those produced by the sulfur-coordinated heme iron of ferric cytochrome P-450, a protein that catalyzes a similar oxygen-dependent reaction. Sulfur 170-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 6185486-10 1983 Although iron bleomycin does not have a polyaromatic structure like heme, many features of its electronic structure at the iron are very similar to those produced by the sulfur-coordinated heme iron of ferric cytochrome P-450, a protein that catalyzes a similar oxygen-dependent reaction. Heme 189-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 6185486-10 1983 Although iron bleomycin does not have a polyaromatic structure like heme, many features of its electronic structure at the iron are very similar to those produced by the sulfur-coordinated heme iron of ferric cytochrome P-450, a protein that catalyzes a similar oxygen-dependent reaction. Iron 123-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 6185486-10 1983 Although iron bleomycin does not have a polyaromatic structure like heme, many features of its electronic structure at the iron are very similar to those produced by the sulfur-coordinated heme iron of ferric cytochrome P-450, a protein that catalyzes a similar oxygen-dependent reaction. Oxygen 262-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 6130930-4 1983 Cimetidine binds reversibly to the hepatic cytochrome P-450 and P-448 systems, resulting in decreased metabolism of drugs that undergo Phase I reactions (e.g., dealkylation and hydroxylation). Cimetidine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-69 6624542-6 1983 HDL-C, A-I and the HDL-C/T-C ratio were directly proportional to liver phospholipids, protein and cytochrome P-450, inversely related to hepatic triglycerides. Triglycerides 145-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 6848512-0 1983 Chiral orientation of prosthetic heme in the cytochrome P-450 active site. Heme 33-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 6848512-3 1983 The orientation of the heme in cytochrome P-450, therefore, is unknown because a crystallographic structure is not available for any form of this enzyme. Heme 23-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 6848512-4 1983 We report here that the absolute configurations of the N-ethylprotoporphyrin IX adducts formed from the prosthetic hemes of cytochrome P-450 and hemoglobin during catalytic turnover of appropriate substrates are identical. N-ethylprotoporphyrin IX 55-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 6848512-5 1983 The prosthetic heme in the inactivated cytochrome P-450 enzyme, therefore, has exactly the same orientation, relative to the fifth iron ligand, as the heme in hemoglobin. Heme 15-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 6848512-5 1983 The prosthetic heme in the inactivated cytochrome P-450 enzyme, therefore, has exactly the same orientation, relative to the fifth iron ligand, as the heme in hemoglobin. Iron 131-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 6848512-5 1983 The prosthetic heme in the inactivated cytochrome P-450 enzyme, therefore, has exactly the same orientation, relative to the fifth iron ligand, as the heme in hemoglobin. Heme 151-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 6848512-6 1983 The approach described here can be used to determine the prosthetic heme orientation in other hemoproteins, including other cytochrome P-450 isozymes, for which x-ray structures are not available. Heme 68-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 6299293-0 1983 Mestranol-induced hypertension: characterization of cytochrome P-450 dependent catechol estrogen formation in brain microsomes. Mestranol 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 6416251-2 1983 Model treatment of the NADPH-dependent first electron transfer reaction between cytochrome P-450 reductase and cytochrome P-450 LM2 in solution. NADP 23-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 6416251-2 1983 Model treatment of the NADPH-dependent first electron transfer reaction between cytochrome P-450 reductase and cytochrome P-450 LM2 in solution. NADP 23-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 6299293-0 1983 Mestranol-induced hypertension: characterization of cytochrome P-450 dependent catechol estrogen formation in brain microsomes. catechol 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 6132787-1 1983 Metabolism of Aromatic hydrocarbons by the cytochrome P-450 system and epoxide hydrolase. Hydrocarbons, Aromatic 14-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 6861274-0 1983 Kinetic evidence for the involvement of multiple forms of human liver cytochrome P-450 in the metabolism of acetylaminofluorene. 2-Acetylaminofluorene 108-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 6418502-10 1983 Another factor which may be involved is heme oxygenase activity, which is markedly induced in the liver after GSH depletion, after cobalt administration (which also depresses cytochrome P-450 activity), and during incubation of isolated hepatocytes. Cobalt 131-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-191 6418502-1 1983 It is apparent that hepatic GSH may function in drug metabolism not only as a substrate for conjugation but also in regulation of cytochrome P-450 activity. Glutathione 28-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 6418502-11 1983 This enzyme catalyzes the rate-limiting step in heme breakdown and may contribute to the loss of cytochrome P-450 activity associated with GSH depletion. Heme 48-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 6418502-8 1983 Ongoing investigations include attempts to identify the cytochrome P-450 isozyme(s) which inhibit this response to GSH depletion. Glutathione 115-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 6418502-9 1983 GSH-lipid peroxidation relationships have already been reported with isolated hepatocytes, and there may be a possible connection between this and the relative instability of cytochrome P-450 in cultured hepatocytes. Glutathione 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-191 6418502-11 1983 This enzyme catalyzes the rate-limiting step in heme breakdown and may contribute to the loss of cytochrome P-450 activity associated with GSH depletion. Glutathione 139-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 6678827-3 1983 Although the mechanism of this pharmacological effect remains unclarified, these results suggest that there is considerable potential for interactions to occur with other drugs metabolized by the same hepatic pathways (cytochrome P 450) in patients undergoing PUVA therapy. puva 260-264 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 219-235 6140286-13 1983 Differences between cimetidine and ranitidine on drug metabolism appear to exist both because the cytochrome P-450 binding affinity for ranitidine is about 10 times lower than cimetidine and because the daily ranitidine dose is 1/4 that of cimetidine. Cimetidine 20-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 6140286-13 1983 Differences between cimetidine and ranitidine on drug metabolism appear to exist both because the cytochrome P-450 binding affinity for ranitidine is about 10 times lower than cimetidine and because the daily ranitidine dose is 1/4 that of cimetidine. Ranitidine 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 6140286-13 1983 Differences between cimetidine and ranitidine on drug metabolism appear to exist both because the cytochrome P-450 binding affinity for ranitidine is about 10 times lower than cimetidine and because the daily ranitidine dose is 1/4 that of cimetidine. Ranitidine 136-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 6140286-13 1983 Differences between cimetidine and ranitidine on drug metabolism appear to exist both because the cytochrome P-450 binding affinity for ranitidine is about 10 times lower than cimetidine and because the daily ranitidine dose is 1/4 that of cimetidine. Ranitidine 136-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 6353196-1 1983 Liver endoplasmic reticulum contains as NADPH-dependent electron transport complex where the family of hemeproteins, termed cytochrome P-450, serve as catalysts for the oxidation of a variety of different organic chemicals. NADP 40-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 6341732-1 1983 The metabolism of tertiary amines is mediated primarily by cytochrome P-450 and MFAO, leading to alpha-C oxidation and N-oxidation, respectively. Amines 27-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-84 6353196-2 1983 The content and inventory of the types of cytochrome P-450 is readily modified following in vivo treatment of animals with "inducing agents" such as barbiturates, steroids and polycyclic hydrocarbons. Barbiturates 149-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 6341732-3 1983 The proposed oxidation of tertiary amines to iminium ions by cytochrome P-450 may explain the isolation of various intramolecular and cyanide-trapped metabolites. Amines 35-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6353196-2 1983 The content and inventory of the types of cytochrome P-450 is readily modified following in vivo treatment of animals with "inducing agents" such as barbiturates, steroids and polycyclic hydrocarbons. Steroids 163-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 6341732-3 1983 The proposed oxidation of tertiary amines to iminium ions by cytochrome P-450 may explain the isolation of various intramolecular and cyanide-trapped metabolites. iminium 45-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6353196-2 1983 The content and inventory of the types of cytochrome P-450 is readily modified following in vivo treatment of animals with "inducing agents" such as barbiturates, steroids and polycyclic hydrocarbons. Hydrocarbons, Cyclic 176-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 6341732-3 1983 The proposed oxidation of tertiary amines to iminium ions by cytochrome P-450 may explain the isolation of various intramolecular and cyanide-trapped metabolites. Cyanides 134-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6356158-10 1983 Thus despite absence of ADH, ADH- deermice can consume large amounts of ethanol: this is associated with increased BEC, SER proliferation, enhanced MEOS activity and quantitative and qualitative changes of cytochrome P-450. Ethanol 72-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-222 7160374-0 1982 Dynamics of the spin transition in camphor-bound ferric cytochrome P-450 versus temperature, pressure and viscosity. Camphor 35-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 6186523-0 1982 Differential effect of phenobarbital and beta-naphthoflavone on the mRNAs coding for cytochrome P450 and NADPH cytochrome P450 reductase. Phenobarbital 23-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 6186523-0 1982 Differential effect of phenobarbital and beta-naphthoflavone on the mRNAs coding for cytochrome P450 and NADPH cytochrome P450 reductase. beta-Naphthoflavone 41-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 7160374-1 1982 The kinetics and equilibrium of the low-spin (LS) to high-spin (HS) transition in camphor-bound ferric cytochrome P-450 have been measured versus temperature, pressure and viscosity at selected pH and KCl concentration. Potassium Chloride 201-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 7160374-1 1982 The kinetics and equilibrium of the low-spin (LS) to high-spin (HS) transition in camphor-bound ferric cytochrome P-450 have been measured versus temperature, pressure and viscosity at selected pH and KCl concentration. Camphor 82-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 7161599-11 1982 Similarities between the hemin-mediated peroxide-supported reactions reported here, and the cytochrome P-450-mediated peroxide-supported reactions reinforce our earlier contentions that the alkaline hemin system appears to be a good model for the in vivo activation of oxygen by hemoproteins. Peroxides 118-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 7161599-11 1982 Similarities between the hemin-mediated peroxide-supported reactions reported here, and the cytochrome P-450-mediated peroxide-supported reactions reinforce our earlier contentions that the alkaline hemin system appears to be a good model for the in vivo activation of oxygen by hemoproteins. Oxygen 269-275 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 7181876-0 1982 Hematoporphyrin photosensitization of epidermal microsomes results in destruction of cytochrome P-450 and in decreased monooxygenase activities and heme content. Hematoporphyrins 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 7181912-0 1982 Occurrence of aromatic methyl migration (NIH-shift) during oxidation of p-methylanisole by hemin-thiolester complex as a cytochrome P-450 model. methyl radical 23-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 7181912-0 1982 Occurrence of aromatic methyl migration (NIH-shift) during oxidation of p-methylanisole by hemin-thiolester complex as a cytochrome P-450 model. 4-methyl anisole 72-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 7181912-0 1982 Occurrence of aromatic methyl migration (NIH-shift) during oxidation of p-methylanisole by hemin-thiolester complex as a cytochrome P-450 model. hemin-thiolester 91-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 6288676-2 1982 Cytochrome P-450 is destroyed during catalytic oxidation of several 4-substituted 3,5-bis(ethoxycarbonyl)-2,6-dimethyl-1,4-dihydropyridine substrates. 4-substituted 3,5-bis(ethoxycarbonyl)-2,6-dimethyl-1,4-dihydropyridine 68-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6288676-9 1982 Hydrogen peroxide formed in situ can support a part of the cytochrome P-450-catalyzed ethyl radical formation and DDEP-dependent self-inactivation. Hydrogen Peroxide 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 6288676-9 1982 Hydrogen peroxide formed in situ can support a part of the cytochrome P-450-catalyzed ethyl radical formation and DDEP-dependent self-inactivation. Ethyl radical 86-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 6288676-10 1982 The results provide persuasive evidence that oxidation of the nitrogen in DDEP by cytochrome P-450 proceeds in one-electron steps. Nitrogen 62-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 6964386-1 1982 Bacterial cytochrome P-450 induced by camphor (P-450cam) is reconstituted with manganese-protoporphyrin IX, yielding an enzyme that displays unique spectral properties relative to previously characterized manganese-porphyrin systems. Camphor 38-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 7107639-0 1982 Endogenous cysteine ligation in ferric and ferrous cytochrome P-450. Cysteine 11-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 6964386-1 1982 Bacterial cytochrome P-450 induced by camphor (P-450cam) is reconstituted with manganese-protoporphyrin IX, yielding an enzyme that displays unique spectral properties relative to previously characterized manganese-porphyrin systems. manganese protoporphyrin 79-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 6964386-1 1982 Bacterial cytochrome P-450 induced by camphor (P-450cam) is reconstituted with manganese-protoporphyrin IX, yielding an enzyme that displays unique spectral properties relative to previously characterized manganese-porphyrin systems. manganese-porphyrin 205-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 7107639-2 1982 Extended x-ray absorption fine structure spectroscopy has been applied to the elucidation of the structure of the heme iron site of bacterial cytochrome P-450. Heme 114-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-158 7107639-2 1982 Extended x-ray absorption fine structure spectroscopy has been applied to the elucidation of the structure of the heme iron site of bacterial cytochrome P-450. Iron 119-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-158 6814537-0 1982 [Comparative study of the reaction kinetics of cytochrome P-450 reduction by NADPH-cytochrome P-450 reductase and dithionite]. Dithionite 114-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 6816226-0 1982 Participation of cytochrome P-450 in nicotine oxidation. Nicotine 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 6286112-1 1982 Human colon tumor cells (cell line LS174T) retain a cytochrome P-450-containing drug metabolism system capable of hydroxylating polycyclic hydrocarbons and the anticancer drug cyclophosphamide. Hydrocarbons, Cyclic 128-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 6814537-1 1982 The reactions of NADPH- or dithionite-dependent reduction of cytochrome P-450 were studied using a stopped flow technique. NADP 17-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6286112-1 1982 Human colon tumor cells (cell line LS174T) retain a cytochrome P-450-containing drug metabolism system capable of hydroxylating polycyclic hydrocarbons and the anticancer drug cyclophosphamide. Cyclophosphamide 176-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 6286112-4 1982 Cytochrome P-450 specific content is increased 2- to 3-fold by treatment with phenobarbital plus hydrocortisone and benz(a)anthracene, respectively. Phenobarbital 78-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6286112-4 1982 Cytochrome P-450 specific content is increased 2- to 3-fold by treatment with phenobarbital plus hydrocortisone and benz(a)anthracene, respectively. Hydrocortisone 97-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6814537-1 1982 The reactions of NADPH- or dithionite-dependent reduction of cytochrome P-450 were studied using a stopped flow technique. Dithionite 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6286112-4 1982 Cytochrome P-450 specific content is increased 2- to 3-fold by treatment with phenobarbital plus hydrocortisone and benz(a)anthracene, respectively. benz(a)anthracene 116-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6814537-4 1982 On the contrary, the breaks on the corresponding plots for the slow phase rate constants are observed at 22 and 33 degrees C for cytochrome P-450 reduction by dithionite and at 31 degrees C for NADPH-dependent reduction of cytochrome P-450. Dithionite 159-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 6814537-4 1982 On the contrary, the breaks on the corresponding plots for the slow phase rate constants are observed at 22 and 33 degrees C for cytochrome P-450 reduction by dithionite and at 31 degrees C for NADPH-dependent reduction of cytochrome P-450. NADP 194-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-239 6814537-5 1982 The coincidence of the values of the rate constants and activation energy (56 +/- 5 kJ/mol) for the fast phase of NADPH-dependent reduction of cytochrome P-450 with values of catalytic constants and activation energy for demethylation of tertiary amines suggests that the first electron transfer process from NADPH-cytochrome P-450 reductase to cytochrome P-450 may be the rate-limiting step. NADP 114-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 6814537-5 1982 The coincidence of the values of the rate constants and activation energy (56 +/- 5 kJ/mol) for the fast phase of NADPH-dependent reduction of cytochrome P-450 with values of catalytic constants and activation energy for demethylation of tertiary amines suggests that the first electron transfer process from NADPH-cytochrome P-450 reductase to cytochrome P-450 may be the rate-limiting step. NADP 114-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 315-331 6814537-5 1982 The coincidence of the values of the rate constants and activation energy (56 +/- 5 kJ/mol) for the fast phase of NADPH-dependent reduction of cytochrome P-450 with values of catalytic constants and activation energy for demethylation of tertiary amines suggests that the first electron transfer process from NADPH-cytochrome P-450 reductase to cytochrome P-450 may be the rate-limiting step. Amines 247-253 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 7108267-9 1982 These studies indicate that orally administered 8-MOP induces hepatic drug-metabolizing enzymes and cytochrome P-450 to a lesser extent than do the barbituates and suggest that this drug could influence the rate of biotransformation of concomitantly administered drugs in patients undergoing PUVA therapy. Methoxsalen 48-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 6292686-3 1982 The formation of this species is inhibited by carbon monoxide and metyrapone, suggesting the involvement of cytochrome P-450. Carbon Monoxide 46-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 6292686-3 1982 The formation of this species is inhibited by carbon monoxide and metyrapone, suggesting the involvement of cytochrome P-450. Metyrapone 66-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 7083192-3 1982 Aminoglutethimide binds competitively to cytochrome P-450 and inhibits a number of steroid hydroxylations but is more active as an aromatase inhibitor. Aminoglutethimide 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 6897045-0 1982 Inactivation of cytochrome P-450 and production of N-alkylated porphyrins caused in isolated hepatocytes by substituted dihydropyridines. Dihydropyridines 120-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 7138513-0 1982 Modification of the cysteine residues of cytochrome P-450cam with 2-bromoacetamido-4-nitrophenol. Cysteine 20-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 7138513-0 1982 Modification of the cysteine residues of cytochrome P-450cam with 2-bromoacetamido-4-nitrophenol. 2-bromoacetamide-4-nitrophenol 66-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 7117257-0 1982 The D(V/K) isotope effect of the cytochrome P-450-mediated oxidation of ethanol and its biological applications. Ethanol 72-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 7161848-6 1982 The relative importance of the FAD-containing monooxygenase vis-a-vis the cytochrome P-450-dependent monooxygenase system is discussed, based on in vitro studies on purified enzymes. Flavin-Adenine Dinucleotide 31-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 7126232-0 1982 Suicidal inactivation of hepatic cytochrome P-450 in vitro by some aliphatic olefins. aliphatic olefins 67-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 6285973-2 1982 The rate of transfer of spin-labeled phospholipid from donor vesicles of sonicated 1-acyl-2-(10-doxylstearoyl)-sn-glycero-3-phosphocholine to other vesicle was determined as a function of content of cytochrome P-450 and the phosphatidylcholine/phosphatidylethanolamine ratio in the acceptor vesicles. Phospholipids 37-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 199-215 6285973-2 1982 The rate of transfer of spin-labeled phospholipid from donor vesicles of sonicated 1-acyl-2-(10-doxylstearoyl)-sn-glycero-3-phosphocholine to other vesicle was determined as a function of content of cytochrome P-450 and the phosphatidylcholine/phosphatidylethanolamine ratio in the acceptor vesicles. 1-acyl-2-(10-doxylstearoyl)-sn-glycero-3-phosphocholine 83-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 199-215 7126360-4 1982 Single and repeated doses of trans-1,2-DCE induced liver aminopyrine demethylase and cytochrome P-450 whereas the cis- isomer decreased them. TRANS-1,2-DICHLOROETHYLENE 29-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 7122438-6 1982 The approaches to the modelling of the cytochrome P-450 catalytic activity with the aid of ferroporphyrine are proposed. ferroporphyrine 91-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 7122439-6 1982 In H2O2-dependent reactions the complex displayed V M and K M similar to those of liver microsomal cytochrome P-450. Hydrogen Peroxide 3-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 6180744-0 1982 Sequential administrations of polyriboinosinic acid and polyribocytidylic acid induce interferon and depress the hepatic cytochrome P-450-dependent monooxygenase system. Polyinosinic acid 30-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 6180744-0 1982 Sequential administrations of polyriboinosinic acid and polyribocytidylic acid induce interferon and depress the hepatic cytochrome P-450-dependent monooxygenase system. Poly C 56-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 7122439-7 1982 In NADPH-dependent reactions the specific activity of the complex was only 1-3% that of cytochrome P-450. NADP 3-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 6810899-6 1982 A reconstituted enzyme system consisting of NADPH, NADPH-cytochrome P-450 reductase, phospholipid and cytochrome P-450 converted mitomycin C to a reactive metabolite(s) under hypoxic conditions. Mitomycin 129-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 6810899-8 1982 These findings support the concept that the cytochrome P-450 system is capable of activating mitomycin C under hypoxic conditions to the alkylating metabolite(s) that is responsible for antineoplastic activity. Mitomycin 93-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-60 7115427-0 1982 The 1,2-dichloroethylenes: their metabolism by hepatic cytochrome p-450 in vitro. 1,2-dichloroethylene 4-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 6980417-1 1982 Cytochrome P-450 (P-450)-dependent aryl hydrocarbon hydroxylase (AHHase) and 7-ethoxycoumarin deethylase (ECDEtase) in human tissues were differentially inhibited by monoclonal antibodies (MAbs) that were prepared to inhibit and completely inhibited the activity of 3-methylcholanthrene-induced rat liver P-450. Methylcholanthrene 266-286 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 7115427-1 1982 Cis- and trans-1,1-dichloroethylene bound to the active site of hepatic microsomal cytochrome P-450 with the production of a Type I difference spectrum and stimulated CO-inhibitable hepatic microsomal NADPH oxidation. cis- and trans-1,1-dichloroethylene 0-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 7115427-1 1982 Cis- and trans-1,1-dichloroethylene bound to the active site of hepatic microsomal cytochrome P-450 with the production of a Type I difference spectrum and stimulated CO-inhibitable hepatic microsomal NADPH oxidation. NADP 201-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 7115427-2 1982 Incubation of cis- and trans-1,2-dichloroethylene plus hepatic microsomes, NADPH-generating system-EDTA resulted in the production of measurable levels of 2,2-dichloroethanol and dichloroacetaldehyde but not of 2-chloroethanol, chloroacetaldehyde or chloroacetic acid and, also, resulted in decreased levels of hepatic microsomal cytochrome P-450 and heme. cis- and trans-1,2-dichloroethylene 14-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 330-355 7115427-2 1982 Incubation of cis- and trans-1,2-dichloroethylene plus hepatic microsomes, NADPH-generating system-EDTA resulted in the production of measurable levels of 2,2-dichloroethanol and dichloroacetaldehyde but not of 2-chloroethanol, chloroacetaldehyde or chloroacetic acid and, also, resulted in decreased levels of hepatic microsomal cytochrome P-450 and heme. NADP 75-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 330-355 7115427-2 1982 Incubation of cis- and trans-1,2-dichloroethylene plus hepatic microsomes, NADPH-generating system-EDTA resulted in the production of measurable levels of 2,2-dichloroethanol and dichloroacetaldehyde but not of 2-chloroethanol, chloroacetaldehyde or chloroacetic acid and, also, resulted in decreased levels of hepatic microsomal cytochrome P-450 and heme. Edetic Acid 99-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 330-355 7115427-2 1982 Incubation of cis- and trans-1,2-dichloroethylene plus hepatic microsomes, NADPH-generating system-EDTA resulted in the production of measurable levels of 2,2-dichloroethanol and dichloroacetaldehyde but not of 2-chloroethanol, chloroacetaldehyde or chloroacetic acid and, also, resulted in decreased levels of hepatic microsomal cytochrome P-450 and heme. 2,2-dichloroethanol 155-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 330-355 7115427-2 1982 Incubation of cis- and trans-1,2-dichloroethylene plus hepatic microsomes, NADPH-generating system-EDTA resulted in the production of measurable levels of 2,2-dichloroethanol and dichloroacetaldehyde but not of 2-chloroethanol, chloroacetaldehyde or chloroacetic acid and, also, resulted in decreased levels of hepatic microsomal cytochrome P-450 and heme. dichloroacetaldehyde 179-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 330-355 7115427-6 1982 The binding and metabolism of the 1,2-dichloroethylenes and the 1,2-dichloroethylene-mediated inactivation of cytochrome P-450 were enhanced per mg of microsomal protein, but generally not per nmole of cytochrome P-450 by prior induction with beta-naphthoflavone or phenobarbital. 1,2-dichloroethylene 34-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 7115427-6 1982 The binding and metabolism of the 1,2-dichloroethylenes and the 1,2-dichloroethylene-mediated inactivation of cytochrome P-450 were enhanced per mg of microsomal protein, but generally not per nmole of cytochrome P-450 by prior induction with beta-naphthoflavone or phenobarbital. 1,2-dichloroethylene 34-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 7115427-6 1982 The binding and metabolism of the 1,2-dichloroethylenes and the 1,2-dichloroethylene-mediated inactivation of cytochrome P-450 were enhanced per mg of microsomal protein, but generally not per nmole of cytochrome P-450 by prior induction with beta-naphthoflavone or phenobarbital. 1,2-dichloroethylene 34-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 7115427-6 1982 The binding and metabolism of the 1,2-dichloroethylenes and the 1,2-dichloroethylene-mediated inactivation of cytochrome P-450 were enhanced per mg of microsomal protein, but generally not per nmole of cytochrome P-450 by prior induction with beta-naphthoflavone or phenobarbital. beta-Naphthoflavone 243-262 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 7115427-6 1982 The binding and metabolism of the 1,2-dichloroethylenes and the 1,2-dichloroethylene-mediated inactivation of cytochrome P-450 were enhanced per mg of microsomal protein, but generally not per nmole of cytochrome P-450 by prior induction with beta-naphthoflavone or phenobarbital. Phenobarbital 266-279 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 7115427-7 1982 It is concluded that multiple forms of hepatic microsomal cytochrome P-450 bind and metabolize the 1,2-dichloroethylenes. 1,2-dichloroethylene 99-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 7115427-8 1982 The role of cytochrome P-450 in the metabolic activation of the dichloroethylenes is considered. Dichloroethylenes 64-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 7106107-2 1982 A study using laser flash photolysis of the dynamics of the reduction of bacterial cytochrome P-450 in the presence of carbon monoxide. Carbon Monoxide 119-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 7106107-4 1982 The intramolecular photoreduction of the bacterial cytochrome P-450 demonstrated by the formation of the complex of reduced cytochrome m with camphor and CO has been studied by photolysis with a laser flash at 265 nm. Camphor 142-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 6279597-0 1982 Low temperature EPR spectroscopic characterization of the interaction of cytochrome P-450cam with a spin label analog of metyrapone. Metyrapone 121-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 6285925-0 1982 Aniline is hydroxylated by the cytochrome P-450-dependent hydroxyl radical-mediated oxygenation mechanism. aniline 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 6285925-0 1982 Aniline is hydroxylated by the cytochrome P-450-dependent hydroxyl radical-mediated oxygenation mechanism. Hydroxyl Radical 58-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 6284506-0 1982 The pressure dependence of the spin equilibrium in camphor-bound ferric cytochrome P-450. Camphor 51-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6284506-1 1982 The spin equilibrium of camphor-bound ferric cytochrome P-450 has been measured between 1-1000 bar (10(5)-10(8) Pa). Camphor 24-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 6284506-7 1982 The correlation can be approximated as delta V = 36 + 18 log K1, which implies that there is a pressure, 3000 bar, for camphor-bound ferric cytochrome P-450 at 4 degrees C, at which the changes in delta V are compensated by the other thermodynamic parameters leaving Ke independent of the solvent conditions. Camphor 119-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 6284506-9 1982 Camphor-bound cytochrome P-450 appears to be a rather flexible protein, having a low denaturing pressure, a large volume change, and a high sensitivity to the protein environment. Camphor 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 6802841-1 1982 A protein-protein association of cytochrome P-450 LM2 with NADPH-cytochrome P-450 reductase, with cytochrome b5, and with both proteins was demonstrated in reconstituted phospholipid vesicles by magnetic circular dichroism difference spectra. Phospholipids 170-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 6802841-6 1982 Upon incorporation of cytochrome b5 into cytochrome P-450- and cytochrome P-450 reductase-containing vesicles, an increase of benzphetamine N-demethylation activity was observed. Benzphetamine 126-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 6802841-6 1982 Upon incorporation of cytochrome b5 into cytochrome P-450- and cytochrome P-450 reductase-containing vesicles, an increase of benzphetamine N-demethylation activity was observed. Benzphetamine 126-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 7093199-11 1982 With sulindac sulfide as substrate, though, both control and phenobarbital-induced microsomes catalyze sulfoxidation to yield the same (+)-sulfoxide enantiomer generated by the purified FAD-containing monoxygenase, suggesting a low degree of participation by the cytochrome P-450 isozymes in sulfoxidation of this compound. sulindac sulfide 5-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 263-279 7093199-11 1982 With sulindac sulfide as substrate, though, both control and phenobarbital-induced microsomes catalyze sulfoxidation to yield the same (+)-sulfoxide enantiomer generated by the purified FAD-containing monoxygenase, suggesting a low degree of participation by the cytochrome P-450 isozymes in sulfoxidation of this compound. Phenobarbital 61-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 263-279 7113709-8 1982 Strong correlations (r greater than 0.91) were seen only between aminopyrine or ethylmorphine demethylase activity and cytochrome P-450 content in samples obtained within 4 hours of death. Aminopyrine 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 6980648-0 1982 Formation of an inactive cytochrome P-450 Fe(II)-metabolite complex after administration of troleandomycin in humans. ammonium ferrous sulfate 42-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 6980648-7 1982 We conclude that repeated administration of troleandomycin induces microsomal enzymes, produces an inactive cytochrome P-450 Fe(II)-metabolite complex, and decreases the clearance of antipyrine in humans. Troleandomycin 44-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 6980648-0 1982 Formation of an inactive cytochrome P-450 Fe(II)-metabolite complex after administration of troleandomycin in humans. Troleandomycin 92-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 6980648-7 1982 We conclude that repeated administration of troleandomycin induces microsomal enzymes, produces an inactive cytochrome P-450 Fe(II)-metabolite complex, and decreases the clearance of antipyrine in humans. ammonium ferrous sulfate 125-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 6980648-1 1982 In rats, it has been shown that troleandomycin induces its own transformation into a metabolite forming an inactive complex with reduced cytochrome P-450. Troleandomycin 32-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-153 6980648-3 1982 In the treated patients, NADPH-cytochrome c reductase activity was increased by 48%; total cytochrome P-450 concentration was also increased, but 33% of total cytochrome P-450 was complexed by a troleandomycin metabolite. Troleandomycin 195-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-175 6980648-4 1982 The cytochrome P-450 Fe(II)-metabolite complex exhibited properties identical to those of the inactive complex formed in rats: it exhibited a Soret peak at 456 nm, was unable to bind CO, and was destroyed by addition of 50 microM potassium ferricyanide. ammonium ferrous sulfate 21-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 6980648-4 1982 The cytochrome P-450 Fe(II)-metabolite complex exhibited properties identical to those of the inactive complex formed in rats: it exhibited a Soret peak at 456 nm, was unable to bind CO, and was destroyed by addition of 50 microM potassium ferricyanide. potassium ferricyanide 230-252 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 6802829-3 1982 A 200% stimulation of N-demethylation by cytochrome b5 was obtained at cytochrome P-450 reductase:cytochrome P-450 ratios similar to those in microsomes, compared to only a 20% stimulation at a ratio of 1:1. Nitrogen 22-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 6802829-3 1982 A 200% stimulation of N-demethylation by cytochrome b5 was obtained at cytochrome P-450 reductase:cytochrome P-450 ratios similar to those in microsomes, compared to only a 20% stimulation at a ratio of 1:1. Nitrogen 22-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 6802829-7 1982 The function of cytochrome b5 was interpreted in terms of a model in which inhibition of cytochrome P-450-mediated reactions results from changes in phospholipid-protein interactions and activation occurs via facilitation of electron transfer between NADPH-cytochrome P-450 reductase and cytochrome P-450 in the membrane. Phospholipids 149-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 6954546-3 1982 A single dose of cobalt-heme (125 mumol/kg of body weight) decreased within 48 hr hepatic cytochrome P-450 to approximately 20% of normal, at which level it remained for 10 days; normal levels were not achieved by 36 days. cobaltiprotoporphyrin 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 7103956-0 1982 Limitations on the metyrapone assay for the major phenobarbital inducible form of cytochrome P-450. Metyrapone 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 7103956-0 1982 Limitations on the metyrapone assay for the major phenobarbital inducible form of cytochrome P-450. Phenobarbital 50-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 6285672-0 1982 Interindividual variation in benzo(a)pyrene metabolism and composition of isoenzymes of cytochrome P-450 as revealed by SDS-gel electrophoresis of human liver microsomal fractions. Benzo(a)pyrene 29-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 6285672-0 1982 Interindividual variation in benzo(a)pyrene metabolism and composition of isoenzymes of cytochrome P-450 as revealed by SDS-gel electrophoresis of human liver microsomal fractions. Sodium Dodecyl Sulfate 120-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 6285672-4 1982 One type of human liver microsomal fraction was selectively induced in specimens obtained from patients with known regular drug intake before death and correlated well in molecular weight to the phenobarbital-inducible form of cytochrome P-450 in the rabbit. Phenobarbital 195-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 227-243 6954546-4 1982 Periodic administration (total, six injections) of a smaller dose of cobalt-heme (50 mumol/kg of body weight) maintained the cytochrome P-450 content at levels approximately 15% of normal for greater than 90 days with concurrent profound impairment of mono-oxygenase reactions catalyzed by this heme protein. cobaltiprotoporphyrin 69-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 6954546-4 1982 Periodic administration (total, six injections) of a smaller dose of cobalt-heme (50 mumol/kg of body weight) maintained the cytochrome P-450 content at levels approximately 15% of normal for greater than 90 days with concurrent profound impairment of mono-oxygenase reactions catalyzed by this heme protein. Heme 76-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 6954546-5 1982 The ability of cobalt-heme to produce profound and prolonged depletion of cytochrome P-450 in vivo provides a valuable model for examining the role of cytochrome P-450-dependent metabolism in the biology of endogenous and exogenous chemicals. cobaltiprotoporphyrin 15-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 6954546-5 1982 The ability of cobalt-heme to produce profound and prolonged depletion of cytochrome P-450 in vivo provides a valuable model for examining the role of cytochrome P-450-dependent metabolism in the biology of endogenous and exogenous chemicals. cobaltiprotoporphyrin 15-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 7074090-0 1982 Kinetics of hepatic cytochrome P-450 reduction: correlation with spin state of the ferric heme. ferric heme 83-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 6803794-0 1982 Novel epoxides formed during the liver cytochrome P-450 oxidation of arachidonic acid. Epoxy Compounds 6-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 7074051-0 1982 Oxidation of trichloroethylene by liver microsomal cytochrome P-450: evidence for chlorine migration in a transition state not involving trichloroethylene oxide. Trichloroethylene 13-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 7074051-0 1982 Oxidation of trichloroethylene by liver microsomal cytochrome P-450: evidence for chlorine migration in a transition state not involving trichloroethylene oxide. Chlorine 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 7074051-1 1982 Trichloroethylene (TCE) was metabolized by cytochrome P-450 containing mixed-function oxidase systems to chloral (2,2,2,-trichloroacetaldehyde), glyoxylic acid, formic acid, CO, and TCE oxide. Trichloroethylene 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 7074051-1 1982 Trichloroethylene (TCE) was metabolized by cytochrome P-450 containing mixed-function oxidase systems to chloral (2,2,2,-trichloroacetaldehyde), glyoxylic acid, formic acid, CO, and TCE oxide. Trichloroethylene 19-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 7074051-1 1982 Trichloroethylene (TCE) was metabolized by cytochrome P-450 containing mixed-function oxidase systems to chloral (2,2,2,-trichloroacetaldehyde), glyoxylic acid, formic acid, CO, and TCE oxide. trichloroacetaldehyde 105-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 7074051-1 1982 Trichloroethylene (TCE) was metabolized by cytochrome P-450 containing mixed-function oxidase systems to chloral (2,2,2,-trichloroacetaldehyde), glyoxylic acid, formic acid, CO, and TCE oxide. trichloroacetaldehyde 114-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 7074051-1 1982 Trichloroethylene (TCE) was metabolized by cytochrome P-450 containing mixed-function oxidase systems to chloral (2,2,2,-trichloroacetaldehyde), glyoxylic acid, formic acid, CO, and TCE oxide. glyoxylic acid 145-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 7074051-1 1982 Trichloroethylene (TCE) was metabolized by cytochrome P-450 containing mixed-function oxidase systems to chloral (2,2,2,-trichloroacetaldehyde), glyoxylic acid, formic acid, CO, and TCE oxide. formic acid 161-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 7074051-1 1982 Trichloroethylene (TCE) was metabolized by cytochrome P-450 containing mixed-function oxidase systems to chloral (2,2,2,-trichloroacetaldehyde), glyoxylic acid, formic acid, CO, and TCE oxide. Carbon Monoxide 174-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 7074051-1 1982 Trichloroethylene (TCE) was metabolized by cytochrome P-450 containing mixed-function oxidase systems to chloral (2,2,2,-trichloroacetaldehyde), glyoxylic acid, formic acid, CO, and TCE oxide. trichloroepoxyethane 182-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 7074051-10 1982 The maximum levels of TCE oxide detected in systems using microsomal fractions and purified cytochrome P-450 were 5-28-fold lower than those predicted from the model. trichloroepoxyethane 22-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 7074051-11 1982 The kinetic data and the discrepancies between the observed metabolites and TCE oxide breakdown products support the view that the epoxide is not an obligate intermediate in the formation of chloral, and an alternative model is presented in which chlorine migration occurs in an oxygenated TCE-cytochrome P-450 transition state. Chlorine 247-255 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-310 6803794-0 1982 Novel epoxides formed during the liver cytochrome P-450 oxidation of arachidonic acid. Arachidonic Acid 69-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 6803786-0 1982 Mechanisms of hydroxylation by cytochrome P-450: exchange of iron-oxygen intermediates with water. Iron 61-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 6803786-0 1982 Mechanisms of hydroxylation by cytochrome P-450: exchange of iron-oxygen intermediates with water. Oxygen 66-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 6803786-0 1982 Mechanisms of hydroxylation by cytochrome P-450: exchange of iron-oxygen intermediates with water. Water 92-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 7054190-0 1982 Effects of 3-methylcholanthrene, beta-naphthoflavone, and phenobarbital on the 3-methylcholanthrene-inducible isozyme of cytochrome P-450 within centrilobular, midzonal, and periportal hepatocytes. Methylcholanthrene 11-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 7054190-0 1982 Effects of 3-methylcholanthrene, beta-naphthoflavone, and phenobarbital on the 3-methylcholanthrene-inducible isozyme of cytochrome P-450 within centrilobular, midzonal, and periportal hepatocytes. beta-Naphthoflavone 33-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 7054190-0 1982 Effects of 3-methylcholanthrene, beta-naphthoflavone, and phenobarbital on the 3-methylcholanthrene-inducible isozyme of cytochrome P-450 within centrilobular, midzonal, and periportal hepatocytes. Phenobarbital 58-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 7054190-0 1982 Effects of 3-methylcholanthrene, beta-naphthoflavone, and phenobarbital on the 3-methylcholanthrene-inducible isozyme of cytochrome P-450 within centrilobular, midzonal, and periportal hepatocytes. Methylcholanthrene 79-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 6896277-4 1982 The decrease in cytochrome P-450 content normally associated with metal administration was, however, prevented, indicating that haem oxygenase induction by metals can proceed without the significant labilization of the haem moiety of cytochrome P-450. Metals 66-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 7171721-1 1982 Absorption spectra of highly purified liver microsomal cytochrome P-450 in non-equilibrium states were obtained at 77 K by reduction with trapped electrons, formed by gamma-irradiation of the water-glycerol matrix. Water 192-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 6801662-0 1982 Cytochrome P-450-dependent oxygenation of arachidonic acid to hydroxyicosatetraenoic acids. Arachidonic Acid 42-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6801662-0 1982 Cytochrome P-450-dependent oxygenation of arachidonic acid to hydroxyicosatetraenoic acids. hydroxyicosatetraenoic acids 62-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6801662-6 1982 THe isolation of these isomeric HETEs suggests that cytochrome P-450 may play a role in the oxidative metabolism of arachidonic acid to physiologically and pharmacologically important hydroxylated unsaturated fatty acids. Arachidonic Acid 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 6801662-6 1982 THe isolation of these isomeric HETEs suggests that cytochrome P-450 may play a role in the oxidative metabolism of arachidonic acid to physiologically and pharmacologically important hydroxylated unsaturated fatty acids. Fatty Acids, Unsaturated 197-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 7171721-1 1982 Absorption spectra of highly purified liver microsomal cytochrome P-450 in non-equilibrium states were obtained at 77 K by reduction with trapped electrons, formed by gamma-irradiation of the water-glycerol matrix. Glycerol 198-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 7171721-2 1982 In contrast to the equilibrium form of ferrous cytochrome P-450 with the heme iron in the high-spin state the non-equilibrium ferrous state has a low-spin heme iron. Heme 73-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 7171721-2 1982 In contrast to the equilibrium form of ferrous cytochrome P-450 with the heme iron in the high-spin state the non-equilibrium ferrous state has a low-spin heme iron. Iron 78-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 7171721-2 1982 In contrast to the equilibrium form of ferrous cytochrome P-450 with the heme iron in the high-spin state the non-equilibrium ferrous state has a low-spin heme iron. Heme 155-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 7171721-2 1982 In contrast to the equilibrium form of ferrous cytochrome P-450 with the heme iron in the high-spin state the non-equilibrium ferrous state has a low-spin heme iron. Iron 160-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 7173298-6 1982 Subjects with a normal liver undergoing treatment with enzyme-inducing drugs, such as phenytoin, phenobarbital and primidone, had higher HDL cholesterol, apoproteins A-I and A-II, HDL cholesterol/total cholesterol ratio, cytochrome P-450 and antipyrine clearance rate than subjects not receiving such therapy. Phenytoin 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-237 7044288-2 1982 These toxic effects of thiono-sulfur-containing compounds appear to be at least partially the result of their metabolism to reactive intermediates by the cytochrome P-450-containing monooxygenase enzyme systems. thiono-sulfur 23-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-170 7044288-3 1982 Covalent binding of (atomic) sulfur released in the cytochrome P-450 monooxygenase catalyzed metabolism of certain thiono-sulfur compounds appears to be responsible for the inhibition of monooxygenase activity and the loss of cytochrome P-450 seen on administration of these thiono-sulfur compounds in vivo or incubation with cytochrome P-450 monooxygenase enzymes in vitro. Sulfur 29-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 7044288-3 1982 Covalent binding of (atomic) sulfur released in the cytochrome P-450 monooxygenase catalyzed metabolism of certain thiono-sulfur compounds appears to be responsible for the inhibition of monooxygenase activity and the loss of cytochrome P-450 seen on administration of these thiono-sulfur compounds in vivo or incubation with cytochrome P-450 monooxygenase enzymes in vitro. thiono-sulfur 115-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 7044288-3 1982 Covalent binding of (atomic) sulfur released in the cytochrome P-450 monooxygenase catalyzed metabolism of certain thiono-sulfur compounds appears to be responsible for the inhibition of monooxygenase activity and the loss of cytochrome P-450 seen on administration of these thiono-sulfur compounds in vivo or incubation with cytochrome P-450 monooxygenase enzymes in vitro. thiono-sulfur 275-288 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 7044288-5 1982 The rationale for implicating metabolites of thiono-sulfur compounds other than atomic sulfur in these effects derives from the experiments with thioacetamide and the fact that atomic sulfur is highly reactive and appears to bind predominantly or exclusively to cytochrome P-450. thiono 45-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-278 7044288-5 1982 The rationale for implicating metabolites of thiono-sulfur compounds other than atomic sulfur in these effects derives from the experiments with thioacetamide and the fact that atomic sulfur is highly reactive and appears to bind predominantly or exclusively to cytochrome P-450. Sulfur 52-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-278 7151648-3 1982 Fractions containing cytochrome P-450 from placentas from smokers will catalyze 7,8-benzoflavone-inhibitable activity in reconstituted systems. alpha-naphthoflavone 80-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 7151648-4 1982 SDS polyacryl-amide gel electrophoresis reveals proteins (molecular weight 70,000-40,000) in cytochrome P-450-containing fractions, derived from women who smoke, which are not detected in material from nonsmokers. Sodium Dodecyl Sulfate 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 7151648-4 1982 SDS polyacryl-amide gel electrophoresis reveals proteins (molecular weight 70,000-40,000) in cytochrome P-450-containing fractions, derived from women who smoke, which are not detected in material from nonsmokers. polyacrylamide 4-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 6118314-5 1982 High binding affinity of cimetidine for cytochrome P 450(Ks = 31 micro M) was seen, while no evidence for ranitidine binding to cytochrome P450- was observed. Cimetidine 25-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 7173298-6 1982 Subjects with a normal liver undergoing treatment with enzyme-inducing drugs, such as phenytoin, phenobarbital and primidone, had higher HDL cholesterol, apoproteins A-I and A-II, HDL cholesterol/total cholesterol ratio, cytochrome P-450 and antipyrine clearance rate than subjects not receiving such therapy. Phenobarbital 97-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-237 7173298-6 1982 Subjects with a normal liver undergoing treatment with enzyme-inducing drugs, such as phenytoin, phenobarbital and primidone, had higher HDL cholesterol, apoproteins A-I and A-II, HDL cholesterol/total cholesterol ratio, cytochrome P-450 and antipyrine clearance rate than subjects not receiving such therapy. Primidone 115-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-237 7173298-8 1982 In the entire population, and also in subjects not taking inducing drugs, when considered separately, plasma HDL cholesterol, apoproteins A-I and A-II and the HDL cholesterol/total cholesterol ratio were significantly correlated with cytochrome P-450 concentration. Cholesterol 113-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-250 7173298-8 1982 In the entire population, and also in subjects not taking inducing drugs, when considered separately, plasma HDL cholesterol, apoproteins A-I and A-II and the HDL cholesterol/total cholesterol ratio were significantly correlated with cytochrome P-450 concentration. Cholesterol 163-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-250 7173298-8 1982 In the entire population, and also in subjects not taking inducing drugs, when considered separately, plasma HDL cholesterol, apoproteins A-I and A-II and the HDL cholesterol/total cholesterol ratio were significantly correlated with cytochrome P-450 concentration. Cholesterol 163-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-250 6270145-6 1981 The phenobarbital-mediated induction of cytochrome P-450 hemoprotein(s) and its monooxygenase function were concomitantly diminished by glucose. Phenobarbital 4-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 6300027-1 1982 Complementary (c)DNA clones of phenobarbital-inducible cytochrome P-450 which has a strong activity toward metabolic activation of aflatoxin B1 were identified by a hybridization-arrested translation assay and a positive hybridization-translation assay. Phenobarbital 31-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 6300027-1 1982 Complementary (c)DNA clones of phenobarbital-inducible cytochrome P-450 which has a strong activity toward metabolic activation of aflatoxin B1 were identified by a hybridization-arrested translation assay and a positive hybridization-translation assay. Aflatoxin B1 131-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 6300027-5 1982 Fourteen nucleotide substitutions occur in their 922 overlapping nucleotides and 7 of them result in 6 amino acid replacements, therefore indicating the presence of at least two similar but distinct mRNAs for phenobarbital-inducible cytochrome P-450. Phenobarbital 209-222 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 233-249 6270145-6 1981 The phenobarbital-mediated induction of cytochrome P-450 hemoprotein(s) and its monooxygenase function were concomitantly diminished by glucose. Glucose 136-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 6270145-8 1981 Glucagon and dibutyryl cAMP enhanced the induction of ALA synthase and cytochrome P-450 by phenobarbital and partially counteracted the glucose effect on both enzymes suggesting that the glucose effect may be mediated by changes in cAMP levels. Glucagon 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 6270145-8 1981 Glucagon and dibutyryl cAMP enhanced the induction of ALA synthase and cytochrome P-450 by phenobarbital and partially counteracted the glucose effect on both enzymes suggesting that the glucose effect may be mediated by changes in cAMP levels. Cyclic AMP 23-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 6270145-8 1981 Glucagon and dibutyryl cAMP enhanced the induction of ALA synthase and cytochrome P-450 by phenobarbital and partially counteracted the glucose effect on both enzymes suggesting that the glucose effect may be mediated by changes in cAMP levels. Phenobarbital 91-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 6270145-8 1981 Glucagon and dibutyryl cAMP enhanced the induction of ALA synthase and cytochrome P-450 by phenobarbital and partially counteracted the glucose effect on both enzymes suggesting that the glucose effect may be mediated by changes in cAMP levels. Glucose 187-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 6274987-0 1981 Side chain hydroxylation of C27-steroids and vitamin D3 by a cytochrome P-450 enzyme system isolated from human liver mitochondria. C 27 28-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6274987-0 1981 Side chain hydroxylation of C27-steroids and vitamin D3 by a cytochrome P-450 enzyme system isolated from human liver mitochondria. Steroids 32-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6274987-0 1981 Side chain hydroxylation of C27-steroids and vitamin D3 by a cytochrome P-450 enzyme system isolated from human liver mitochondria. Cholecalciferol 45-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 6274987-1 1981 The present study was undertaken to obtain information on the involvement of cytochrome P-450 in the 26-hydroxylation on bile acid intermediates and in the 25-hydroxylation of vitamin D3 in human liver mitochondria. Bile Acids and Salts 121-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 6274987-7 1981 The cytochrome P-450 preparation catalyzed 26-hydroxylation of C27-steroids and 25-hydroxylation of vitamin D3 when reconstructed with NADPH, the ferredoxin and the ferredoxin reductase. Steroids 67-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 6274987-7 1981 The cytochrome P-450 preparation catalyzed 26-hydroxylation of C27-steroids and 25-hydroxylation of vitamin D3 when reconstructed with NADPH, the ferredoxin and the ferredoxin reductase. Cholecalciferol 100-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 6274987-7 1981 The cytochrome P-450 preparation catalyzed 26-hydroxylation of C27-steroids and 25-hydroxylation of vitamin D3 when reconstructed with NADPH, the ferredoxin and the ferredoxin reductase. NADP 135-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 6274987-11 1981 It is concluded that human liver mitochondria contain cytochrome P-450 involved in the oxidation of the side chain of C27-steroids and vitamin D3. C 27 118-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 6274987-11 1981 It is concluded that human liver mitochondria contain cytochrome P-450 involved in the oxidation of the side chain of C27-steroids and vitamin D3. Steroids 122-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 6274987-11 1981 It is concluded that human liver mitochondria contain cytochrome P-450 involved in the oxidation of the side chain of C27-steroids and vitamin D3. Cholecalciferol 135-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 6457649-0 1981 Comparison of the effects of inhibitors of cytochrome P-450-mediated reactions on human platelet aggregation and arachidonic acid metabolism. Arachidonic Acid 113-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 6797104-1 1981 Phenobarbital stimulates the induction of liver microsomal drug-metabolizing enzyme, namely, cytochrome P-450, which enhances the rate of conversion of FT-207 to 5-FU, the active substance. Phenobarbital 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 6797104-1 1981 Phenobarbital stimulates the induction of liver microsomal drug-metabolizing enzyme, namely, cytochrome P-450, which enhances the rate of conversion of FT-207 to 5-FU, the active substance. Tegafur 152-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 6797104-1 1981 Phenobarbital stimulates the induction of liver microsomal drug-metabolizing enzyme, namely, cytochrome P-450, which enhances the rate of conversion of FT-207 to 5-FU, the active substance. Fluorouracil 162-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 6797104-2 1981 When FT-207 is administered in combination with phenobarbital to cancer patients, the fluctuation in level of the drug-metabolizing enzyme, cytochrome P-450, should be taken into consideration. Phenobarbital 48-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 7295780-2 1981 MCD was applied to ferric and ferrous low-spin complexes of cytochrome P-450 cam to elucidate the electronic states and the nature of the axial ligands of the heme in cytochrome P-450cam. Heme 159-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 7295780-6 1981 The ratio (4.2) for the alpha-picoline-bound form of cytochrome P-450cam, however, was the closest to that (2.7) for the camphor-free form of cytochrome P-450cam among those (4.2-9.0) for the external ligand-bound form of cytochrome P-450cam. Picolines 24-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 7295780-6 1981 The ratio (4.2) for the alpha-picoline-bound form of cytochrome P-450cam, however, was the closest to that (2.7) for the camphor-free form of cytochrome P-450cam among those (4.2-9.0) for the external ligand-bound form of cytochrome P-450cam. Camphor 121-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 7295780-7 1981 The ratio for the 2-methylimidazole-bound form of cytochrome P-450cam was the smallest among those of cytochrome P-450cam bound with imidizole derivatives. 2-methylimidazole 18-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 6457649-1 1981 Metyrapone and SKF-525A, together with amphenone B, a structural analogue of metyrapone, which are all inhibitors of cytochrome P-450-mediated reactions, were shown to inhibit the arachidonic acid-induced aggregation of human platelets. Metyrapone 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 7295780-7 1981 The ratio for the 2-methylimidazole-bound form of cytochrome P-450cam was the smallest among those of cytochrome P-450cam bound with imidizole derivatives. imidizole 133-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 6457649-1 1981 Metyrapone and SKF-525A, together with amphenone B, a structural analogue of metyrapone, which are all inhibitors of cytochrome P-450-mediated reactions, were shown to inhibit the arachidonic acid-induced aggregation of human platelets. Proadifen 15-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 6457649-1 1981 Metyrapone and SKF-525A, together with amphenone B, a structural analogue of metyrapone, which are all inhibitors of cytochrome P-450-mediated reactions, were shown to inhibit the arachidonic acid-induced aggregation of human platelets. amphenone B 39-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 6457649-1 1981 Metyrapone and SKF-525A, together with amphenone B, a structural analogue of metyrapone, which are all inhibitors of cytochrome P-450-mediated reactions, were shown to inhibit the arachidonic acid-induced aggregation of human platelets. Metyrapone 77-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 6457649-1 1981 Metyrapone and SKF-525A, together with amphenone B, a structural analogue of metyrapone, which are all inhibitors of cytochrome P-450-mediated reactions, were shown to inhibit the arachidonic acid-induced aggregation of human platelets. Arachidonic Acid 180-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 6457649-7 1981 These data provide further support for a role of cytochrome P-450 in thromboxane synthesis and platelet aggregation. Thromboxanes 69-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 6797432-0 1981 Ethanol-mediated increase in cytochrome P-450 in cultured hepatocytes. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 6272881-1 1981 The Cu-tyrosine complex, a low molecular weight analog of superoxide dismutase, exerts an inhibiting effect on cytochrome P-450. copper(II)-tyrosine complex 4-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 7285242-4 1981 The binding of type I compounds, hexobarbital and androstanedione, with cytochrome P-450, as determined by the magnitude of the type I spectral change of microsomes, was markedly enhanced at alkaline pH compared to that at acid pH. Hexobarbital 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 6272881-3 1981 In case of p-hydroxylation of aniline the inhibiting effect of the Cu-tyrosine complex is much more pronounced than its inactivating effect on cytochrome P-450. aniline 30-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 6272881-3 1981 In case of p-hydroxylation of aniline the inhibiting effect of the Cu-tyrosine complex is much more pronounced than its inactivating effect on cytochrome P-450. copper(II)-tyrosine complex 67-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 6272881-6 1981 In a soluble system containing isolated cytochrome P-450 and cumole hydroperoxide only the aniline p-hydroxylation reaction was found sensitive to the effect of superoxide dismutase. aniline 91-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 7285242-4 1981 The binding of type I compounds, hexobarbital and androstanedione, with cytochrome P-450, as determined by the magnitude of the type I spectral change of microsomes, was markedly enhanced at alkaline pH compared to that at acid pH. Androstanedione 50-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 7285242-8 1981 On the contrary, the absorbance magnitude between peak and trough in the aniline- or alcohol-induced difference spectrum of microsomes was enhanced by decreasing the pH, indicating easy complex formation of type II and reverse type I compounds with cytochrome P-450 in the acid rather than the alkaline region. aniline 73-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 249-265 7285242-8 1981 On the contrary, the absorbance magnitude between peak and trough in the aniline- or alcohol-induced difference spectrum of microsomes was enhanced by decreasing the pH, indicating easy complex formation of type II and reverse type I compounds with cytochrome P-450 in the acid rather than the alkaline region. Alcohols 85-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 249-265 6796087-0 1981 1-Chloro-2,2,2-trifluoroethyl radical: Formation from halothane by human cytochrome P-450 in reconstituted vesicles and binding to phospholipids. 1-chloro-2,2,2-trifluoroethyl radical 0-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 7295776-2 1981 Reduced P-450SCC at pH 7.4 exhibited the V4 line at 1342 cm-1, which is an unusually low frequency compared with an ordinary protohemoprotein but is common to the family of cytochrome P-450, suggesting the coordination of a strong pi-donor such as thiolate anion at the fifth coordination position of the heme iron. thiolate anion 248-262 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-189 7295776-2 1981 Reduced P-450SCC at pH 7.4 exhibited the V4 line at 1342 cm-1, which is an unusually low frequency compared with an ordinary protohemoprotein but is common to the family of cytochrome P-450, suggesting the coordination of a strong pi-donor such as thiolate anion at the fifth coordination position of the heme iron. Heme 305-309 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-189 7295776-2 1981 Reduced P-450SCC at pH 7.4 exhibited the V4 line at 1342 cm-1, which is an unusually low frequency compared with an ordinary protohemoprotein but is common to the family of cytochrome P-450, suggesting the coordination of a strong pi-donor such as thiolate anion at the fifth coordination position of the heme iron. Iron 310-314 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-189 7295776-6 1981 In the presence of 20alpha-hydroxycholesterol, oxidized P-450SCC gave the V10 line at 1637 cm-1, i.e., at a frequency similar to that of low-spin type cytochrome P-450. 20-hydroxycholesterol 19-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 6796087-0 1981 1-Chloro-2,2,2-trifluoroethyl radical: Formation from halothane by human cytochrome P-450 in reconstituted vesicles and binding to phospholipids. Halothane 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 21043244-8 1981 Studies on the effects of the in vitro modifiers of monooxygenase activity alpha-naphthoflavone and metyrapone further supported the hypothesis that the two phases of O-deethylase activity represent two different forms or populations of cytochrome P-450. alpha-naphthoflavone 75-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 237-253 21043244-8 1981 Studies on the effects of the in vitro modifiers of monooxygenase activity alpha-naphthoflavone and metyrapone further supported the hypothesis that the two phases of O-deethylase activity represent two different forms or populations of cytochrome P-450. Metyrapone 100-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 237-253 6897891-2 1981 It follows that alterations in the activities of the enzymes of heme metabolism are often reflected in the heme dependent cellular functions, particularly those which depend on cytochrome P-450. Heme 64-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 7285236-0 1981 Ellipticines and human liver microsomes: spectral interaction with cytochrome P-450 and hydroxylation. Ellipticines 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 7285236-2 1981 Some pharmacological properties of ellipticine (E) and its derivatives linked to their interaction with cytochrome P-450 have been investigated with human liver microsomes. ellipticine 35-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 7285236-3 1981 9-Hydroxyellipticine (9-OHE) interacts with human liver cytochrome P-450 exhibiting a type II spectrum (lambda max: 428 nm, Ks = 1.1 microM). 9-hydroxyellipticine 0-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 7285236-3 1981 9-Hydroxyellipticine (9-OHE) interacts with human liver cytochrome P-450 exhibiting a type II spectrum (lambda max: 428 nm, Ks = 1.1 microM). 9-hydroxyellipticine 22-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 7297566-12 1981 The monooxygenase activities, the sensitivity to in vitro alpha-naphthoflavone and metyrapone, the results of steroid metabolism, and slab gel electrophoresis are strong indications for multiplicity of human liver cytochrome P-450. alpha-naphthoflavone 58-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-230 7297566-12 1981 The monooxygenase activities, the sensitivity to in vitro alpha-naphthoflavone and metyrapone, the results of steroid metabolism, and slab gel electrophoresis are strong indications for multiplicity of human liver cytochrome P-450. Metyrapone 83-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-230 7297566-12 1981 The monooxygenase activities, the sensitivity to in vitro alpha-naphthoflavone and metyrapone, the results of steroid metabolism, and slab gel electrophoresis are strong indications for multiplicity of human liver cytochrome P-450. Steroids 110-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-230 6897891-2 1981 It follows that alterations in the activities of the enzymes of heme metabolism are often reflected in the heme dependent cellular functions, particularly those which depend on cytochrome P-450. Heme 107-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 6897891-3 1981 Moreover, perturbations in cellular GSH levels may alter the biological inactivation of the intermediates of cytochrome P-450 activity normally inactivated by GSH-conjugation. Glutathione 36-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 6897891-3 1981 Moreover, perturbations in cellular GSH levels may alter the biological inactivation of the intermediates of cytochrome P-450 activity normally inactivated by GSH-conjugation. Glutathione 159-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 7196447-4 1981 The involvement of the cytochrome P-450 system in the activation of 6-thiopurine to the chemically reactive metabolite was supported by the effects of inhibitors of cytochrome P-450 mediated reactions and phenobarbital induction. Mercaptopurine 68-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 7196447-4 1981 The involvement of the cytochrome P-450 system in the activation of 6-thiopurine to the chemically reactive metabolite was supported by the effects of inhibitors of cytochrome P-450 mediated reactions and phenobarbital induction. Mercaptopurine 68-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-181 7196447-4 1981 The involvement of the cytochrome P-450 system in the activation of 6-thiopurine to the chemically reactive metabolite was supported by the effects of inhibitors of cytochrome P-450 mediated reactions and phenobarbital induction. Phenobarbital 205-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 6946467-4 1981 These results suggest that each molecular form of cytochrome P-450 that converts antipyrine to a different metabolite exhibits genetically controlled interindividual variations in activity. Antipyrine 81-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 6272772-0 1981 Glutathione-hemin complex as a cytochrome P-450 model characterization of the complex and its aromatic oxidation activities. glutathione-hemin 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 7271831-0 1981 Cytochrome P-450-dependent metabolism of 1,1,2,2-tetrachloroethane to dichloroacetic acid in vitro. 1,1,2,2-tetrachloroethane 41-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 6114841-0 1981 Is a photo-oxidation product of histidine one of the triggers of induction of cytochrome P-450 drug-metabolizing enzyme systems? Histidine 32-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 7288293-1 1981 Cholesterol 7 alpha-hydroxylase, the rate-limiting enzyme for bile acid synthesis, was shown to be copurified with human liver microsomal cytochrome P-450. Bile Acids and Salts 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-154 7288293-2 1981 When these cytochrome P-450 species were reconstituted in phospholipid-cholesterol vesicles together with NADPH-cytochrome P-450 reductase, high cholesterol 7 alpha-hydroxylase activity was obtained in the presence of NADPH. Phospholipids 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 7288293-2 1981 When these cytochrome P-450 species were reconstituted in phospholipid-cholesterol vesicles together with NADPH-cytochrome P-450 reductase, high cholesterol 7 alpha-hydroxylase activity was obtained in the presence of NADPH. NADP 106-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 7271831-0 1981 Cytochrome P-450-dependent metabolism of 1,1,2,2-tetrachloroethane to dichloroacetic acid in vitro. Dichloroacetic Acid 70-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 7250371-0 1981 Effects of dietary lipid and phenobarbitone on the distribution and concentration of cytochrome P-450 in the liver studied by quantitative cytochemistry. Phenobarbital 29-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 7262071-4 1981 This establishes a role for cytochrome b5 in donating electrons for the reduction of oxy-cytochrome P-450 to the active oxygen complex of cytochrome P-450 but also points to large variations in the importance of this role depending on the experimental conditions, the species of P-450 involved and the substrates employed. Oxygen 120-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 7262071-4 1981 This establishes a role for cytochrome b5 in donating electrons for the reduction of oxy-cytochrome P-450 to the active oxygen complex of cytochrome P-450 but also points to large variations in the importance of this role depending on the experimental conditions, the species of P-450 involved and the substrates employed. Oxygen 120-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-154 6264953-0 1981 Phospholipid interactions with cytochrome P-450 in reconstituted vesicles. Phospholipids 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 7217086-0 1981 Destruction of cytochrome P-450 by ethylene. ethylene 35-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 7217086-2 1981 The prosthetic heme of cytochrome P-450 is converted during the metabolism of ethylene into an abnormal hepatic porphyrin. Heme 15-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 7217086-2 1981 The prosthetic heme of cytochrome P-450 is converted during the metabolism of ethylene into an abnormal hepatic porphyrin. ethylene 78-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 7217086-6 1981 Strong support is provided by the structure of this porphyrin for our contention that the prosthetic heme of cytochrome P-450 is alkylated during attempted transfer of the catalytically-activated oxygen to the pi-bond of destructive unsaturated substrates. Heme 101-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 7217086-6 1981 Strong support is provided by the structure of this porphyrin for our contention that the prosthetic heme of cytochrome P-450 is alkylated during attempted transfer of the catalytically-activated oxygen to the pi-bond of destructive unsaturated substrates. Oxygen 196-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 7296714-0 1981 Oxidative dealkylation of tertiary amines by iron(III) porphyrin-iodosoxylene system as a model of cytochrome P-450. Amines 35-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 6973977-0 1981 Self-induction by triacetyloleandomycin of its own transformation into a metabolite forming a stable 456 nm-absorbing complex with cytochrome P-450. Troleandomycin 18-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 7296714-0 1981 Oxidative dealkylation of tertiary amines by iron(III) porphyrin-iodosoxylene system as a model of cytochrome P-450. iron(iii) porphyrin 45-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 7296714-0 1981 Oxidative dealkylation of tertiary amines by iron(III) porphyrin-iodosoxylene system as a model of cytochrome P-450. iodosoxylene 65-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 7297957-1 1981 Hemoglobin and cytochrome P-450 have in common heme structure (i.e. protoporphyrin (IX), binding ability to molecular oxygen or carbon monoxide and enzyme-like activity (i.e. aniline hydroxylation; J.B.C. Heme 47-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 7297957-1 1981 Hemoglobin and cytochrome P-450 have in common heme structure (i.e. protoporphyrin (IX), binding ability to molecular oxygen or carbon monoxide and enzyme-like activity (i.e. aniline hydroxylation; J.B.C. protoporphyrin IX 68-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 7297957-1 1981 Hemoglobin and cytochrome P-450 have in common heme structure (i.e. protoporphyrin (IX), binding ability to molecular oxygen or carbon monoxide and enzyme-like activity (i.e. aniline hydroxylation; J.B.C. Oxygen 118-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 7297957-1 1981 Hemoglobin and cytochrome P-450 have in common heme structure (i.e. protoporphyrin (IX), binding ability to molecular oxygen or carbon monoxide and enzyme-like activity (i.e. aniline hydroxylation; J.B.C. Carbon Monoxide 128-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 7297957-1 1981 Hemoglobin and cytochrome P-450 have in common heme structure (i.e. protoporphyrin (IX), binding ability to molecular oxygen or carbon monoxide and enzyme-like activity (i.e. aniline hydroxylation; J.B.C. aniline 175-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 6786868-2 1981 Many xenobiotics inhibit steroid hormone production as a result of interactions with cytochrome P-450-containing hydroxylases in adrenal mitochondria or microsomes. Steroids 25-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 7233442-0 1981 Depression of cytochrome P-450-dependent drug biotransformation by adriamycin. Doxorubicin 67-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 6973978-0 1981 Hypoactivity of cytochrome P-450 after triacetyloleandomycin administration. Troleandomycin 39-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 6783456-0 1981 Electrophoretically homogeneous cytochrome P-450: influence of a series of unsaturated fatty acids on the reconstituted hydroxylase activity. Fatty Acids, Unsaturated 75-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 6784732-0 1981 Effects of indole and tryptophan on cytochrome P-450, dimethylnitrosamine demethylase, and arylhydrocarbon hydroxylase activities. indole 11-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-85 6784732-0 1981 Effects of indole and tryptophan on cytochrome P-450, dimethylnitrosamine demethylase, and arylhydrocarbon hydroxylase activities. Tryptophan 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-85 7448777-5 1981 These results suggest that the inhibitory effects of quercetin, morin, and kaempferol on monooxygenase activity may be caused at least in part by an inhibition in the reduction of cytochrome P-450. Quercetin 53-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 6894611-4 1981 Concomitant with heme oxygenase induction by antimony, microsomal heme and cytochrome P-450 contents decreased, the cyto-chrome P-450-dependent mixed function oxidase system was impaired, and delta-ami-nolevulinate synthase (ALAS), the rate-limiting enzyme of heme synthesis, underwent the sequential changes-initial inhibition followed by rebound induction-usually associated with the administration of transition elements such as cobalt. Heme 17-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 7344478-0 1981 Regio- and stereoselectivity of hepatic cytochrome P-450 toward polycyclic aromatic hydrocarbon substrates. Polycyclic Aromatic Hydrocarbons 64-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 7344478-1 1981 Although the cytochrome P-450 system shows broad substrate specificity toward the PAH, it also has regio- and stereoselectivity. Polycyclic Aromatic Hydrocarbons 82-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 7344478-3 1981 Interestingly, the cytochrome P-450 system and epoxide hydrolase favor the formation of dihydrodiols with R,R configuration. trans-1,2-dihydro-1,2-naphthalenediol 88-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 7344490-0 1981 The cytochrome P-450-mediated metabolism of biphenyl and the 4-halobiphenyls. halobiphenyls 63-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 7344518-4 1981 Studies in vitro and in vivo indicate that cytochrome P-450 enzymes in the target tissues mediate the formation of highly reactive, electrophilic furan metabolites that bind covalently to tissue macromolecules. furan 146-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 6124093-0 1981 Reaction schemes for the degradation of cytochrome P-450 by allyl-iso-propylacetamide and fluroxene. Allylisopropylacetamide 60-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 6124093-0 1981 Reaction schemes for the degradation of cytochrome P-450 by allyl-iso-propylacetamide and fluroxene. fluroxene 90-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 7285278-0 1981 Chloroethanes : their metabolism by hepatic cytochrome P-450 in vitro. Ethyl Chloride 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-60 7285278-3 1981 Both NADPH oxidation and metabolite production are inhibited by CO, SKF 525A and/or metyrapone The induction of cytochrome P-450 with phenobarbital enhances the binding and metabolism of the chloroalkanes, while the induction of cytochrome P-448 with beta-naphthoflavone does not. NADP 5-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 7285278-3 1981 Both NADPH oxidation and metabolite production are inhibited by CO, SKF 525A and/or metyrapone The induction of cytochrome P-450 with phenobarbital enhances the binding and metabolism of the chloroalkanes, while the induction of cytochrome P-448 with beta-naphthoflavone does not. Metyrapone 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 7285278-3 1981 Both NADPH oxidation and metabolite production are inhibited by CO, SKF 525A and/or metyrapone The induction of cytochrome P-450 with phenobarbital enhances the binding and metabolism of the chloroalkanes, while the induction of cytochrome P-448 with beta-naphthoflavone does not. Phenobarbital 134-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 7285278-3 1981 Both NADPH oxidation and metabolite production are inhibited by CO, SKF 525A and/or metyrapone The induction of cytochrome P-450 with phenobarbital enhances the binding and metabolism of the chloroalkanes, while the induction of cytochrome P-448 with beta-naphthoflavone does not. chloroalkanes 191-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 7285278-3 1981 Both NADPH oxidation and metabolite production are inhibited by CO, SKF 525A and/or metyrapone The induction of cytochrome P-450 with phenobarbital enhances the binding and metabolism of the chloroalkanes, while the induction of cytochrome P-448 with beta-naphthoflavone does not. beta-Naphthoflavone 251-270 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 7285278-4 1981 1,1,1-trichloroethane is converted to 2,2,2-trichloroethanol by hepatic microsomal cytochrome P-450, while the major metabolites of 1,1,2-trichloroethane and 1,1,2,2-tetrachloroethane from this enzyme system are mono- and dichloroacetate, respectively. 1,1,1-trichloroethane 0-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 7285278-4 1981 1,1,1-trichloroethane is converted to 2,2,2-trichloroethanol by hepatic microsomal cytochrome P-450, while the major metabolites of 1,1,2-trichloroethane and 1,1,2,2-tetrachloroethane from this enzyme system are mono- and dichloroacetate, respectively. 2,2,2-trichloroethanol 38-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 7285278-4 1981 1,1,1-trichloroethane is converted to 2,2,2-trichloroethanol by hepatic microsomal cytochrome P-450, while the major metabolites of 1,1,2-trichloroethane and 1,1,2,2-tetrachloroethane from this enzyme system are mono- and dichloroacetate, respectively. 1,1,2-trichloroethane 132-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 7448777-5 1981 These results suggest that the inhibitory effects of quercetin, morin, and kaempferol on monooxygenase activity may be caused at least in part by an inhibition in the reduction of cytochrome P-450. kaempferol 75-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 7285278-4 1981 1,1,1-trichloroethane is converted to 2,2,2-trichloroethanol by hepatic microsomal cytochrome P-450, while the major metabolites of 1,1,2-trichloroethane and 1,1,2,2-tetrachloroethane from this enzyme system are mono- and dichloroacetate, respectively. 1,1,2,2-tetrachloroethane 158-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 7285278-4 1981 1,1,1-trichloroethane is converted to 2,2,2-trichloroethanol by hepatic microsomal cytochrome P-450, while the major metabolites of 1,1,2-trichloroethane and 1,1,2,2-tetrachloroethane from this enzyme system are mono- and dichloroacetate, respectively. mono- and dichloroacetate 212-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 7251630-6 1981 Dialkylnitrosamines are shown to be activated by oxidative dealkylation via cytochrome P-450 enzyme systems. dialkylnitrosamines 0-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 7308271-5 1981 Alcohol excess was associated with decreased cytochrome P-450 content and AHH activity and this effect was independent of the histological status of the biopsy. Alcohols 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 7341283-9 1981 It is concluded that this provides evidence for the involvement of at least two different forms of cytochrome P-450 in antipyrine metabolism in man. Antipyrine 119-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 7450397-1 1981 The aim of this study was to look for correlations between the metabolic clearance rate of antipyrine and (a) the concentration of cytochrome P-450, and (b) the NADPH cytochrome c reductase, aminopyrine demethylase, and aniline hydroxylase activities in the liver microsomes of 20 patients without liver diseases. Antipyrine 91-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 7267701-6 1981 It is concluded that different species of the drug-oxidizing enzymes (cytochrome P-450 system) are involved in the metabolism of debrisoquine and antipyrine. Debrisoquin 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 7267701-6 1981 It is concluded that different species of the drug-oxidizing enzymes (cytochrome P-450 system) are involved in the metabolism of debrisoquine and antipyrine. Antipyrine 146-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 7256785-2 1981 The increase in 1,8-ANS binding is in accordance with the simultaneous increase of the ethoxycoumarin O-de-ethylase activity and cytochrome P-450 concentration. 1-anilino-8-naphthalenesulfonate 16-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 7256785-6 1981 The data suggest that chronic ethanol administration may effect the biotransformation enzyme activities by changing the structural properties of the membranes as well as increasing the cytochrome P-450 concentration. Ethanol 30-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-201 6894538-3 1980 The decreases in cytochrome P-450 and haem contents and the sequential changes in delta-aminolaevulinate synthase (EC 2.3.1.37) activity that occur concomitant with haem oxygenase induction were largely eliminated with simultaneous or prior treatment with Mn2+ or Zn2+, but not when Mn2+ or Zn2+ was administered after Sn2+ or Ni2+. Manganese(2+) 256-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 6111318-0 1980 Role of hepatic microsomal cytochrome P-450 in the toxicity of fluorinated ether anesthetics. ether anesthetics 75-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 6894538-6 1980 The Zn2+ blockade of Cd2+ induction was examined in detail, and prior or simultaneous administration of Zn2+ was found to be effective in blocking the induction of haem oxygenase and the concomitant decreases in cytochrome P-450 and haem contents, ethylmorphine demethylase activity and the sequential changes in delta-aminolaevulinate synthase activity. Zinc 104-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 212-228 7408405-7 1980 It is suggested that N-demethylation of theophylline to 3MX and 1MU is catalyzed by a different form of cytochrome P-450 than that involved in the 8-hydroxylation to DMU. Theophylline 40-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 7408405-7 1980 It is suggested that N-demethylation of theophylline to 3MX and 1MU is catalyzed by a different form of cytochrome P-450 than that involved in the 8-hydroxylation to DMU. 1,3-dimethyluric acid 166-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 16661486-0 1980 Time Course of Induction of Cytochrome P-450, NADPH-Cytochrome c Reductase, and Cinnamic Acid Hydroxylase by Phenobarbital, Ethanol, Herbicides, and Manganese in Higher Plant Microsomes. Phenobarbital 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 16661486-0 1980 Time Course of Induction of Cytochrome P-450, NADPH-Cytochrome c Reductase, and Cinnamic Acid Hydroxylase by Phenobarbital, Ethanol, Herbicides, and Manganese in Higher Plant Microsomes. Ethanol 124-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 16661486-0 1980 Time Course of Induction of Cytochrome P-450, NADPH-Cytochrome c Reductase, and Cinnamic Acid Hydroxylase by Phenobarbital, Ethanol, Herbicides, and Manganese in Higher Plant Microsomes. Manganese 149-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 16661486-2 1980 Although the microsomal hydroxylating complex is already induced by the slicing and aging process, 25 millimolar MnCl(2), 4 millimolar phenobarbital, and 300 millimolar ethanol caused a marked increase of hydroxylase activity and cytochrome P-450 content and shifted their time course. manganese chloride 113-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-246 16661486-2 1980 Although the microsomal hydroxylating complex is already induced by the slicing and aging process, 25 millimolar MnCl(2), 4 millimolar phenobarbital, and 300 millimolar ethanol caused a marked increase of hydroxylase activity and cytochrome P-450 content and shifted their time course. Phenobarbital 135-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-246 16661486-2 1980 Although the microsomal hydroxylating complex is already induced by the slicing and aging process, 25 millimolar MnCl(2), 4 millimolar phenobarbital, and 300 millimolar ethanol caused a marked increase of hydroxylase activity and cytochrome P-450 content and shifted their time course. Ethanol 169-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-246 6779432-3 1980 In addition the in vitro inhibition of 7-ethoxycoumarin-deethylase by metyrapone (phenobarbital induced cytochrome P-450) or naphthoflavone (benzo[a]pyrene induced cytochrome P-448) was estimated. Metyrapone 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 6773745-2 1980 This activity employs cytochrome P-450 as an oxygen donor. Oxygen 45-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 7437045-0 1980 Iron-porphyrin catalysis of the oxidative dealkylation of para-nitro-anisole and 7-ethoxycoumarin by cumylhydroperoxide: a possible model for the corresponding cytochrome P 450-dependent reactions. iron-porphyrin 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 6779432-3 1980 In addition the in vitro inhibition of 7-ethoxycoumarin-deethylase by metyrapone (phenobarbital induced cytochrome P-450) or naphthoflavone (benzo[a]pyrene induced cytochrome P-448) was estimated. Phenobarbital 82-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 7437045-0 1980 Iron-porphyrin catalysis of the oxidative dealkylation of para-nitro-anisole and 7-ethoxycoumarin by cumylhydroperoxide: a possible model for the corresponding cytochrome P 450-dependent reactions. 4-nitroanisole 58-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 7437045-0 1980 Iron-porphyrin catalysis of the oxidative dealkylation of para-nitro-anisole and 7-ethoxycoumarin by cumylhydroperoxide: a possible model for the corresponding cytochrome P 450-dependent reactions. 7-ethoxycoumarin 81-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 7437045-0 1980 Iron-porphyrin catalysis of the oxidative dealkylation of para-nitro-anisole and 7-ethoxycoumarin by cumylhydroperoxide: a possible model for the corresponding cytochrome P 450-dependent reactions. cumene hydroperoxide 101-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 7391085-0 1980 The kinetics of reduction of cytochrome P-450cam by the dithionite anion monomer. dithionite anion 56-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 7236798-1 1980 This secondary structure of soluble cytochrome P-450 and the one incorporated into liposomes from egg lecithin and microsomal lipids has been studied. Lecithins 102-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 7236798-3 1980 The binding of haemoprotein with the type II substrates--octylamine and diaminooctan, slightly increases alpha-helices in soluble cytochrome P-450. octylamine 57-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 7236798-3 1980 The binding of haemoprotein with the type II substrates--octylamine and diaminooctan, slightly increases alpha-helices in soluble cytochrome P-450. diaminooctan 72-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 7236798-5 1980 Cytochrome P-450 incorporated into the artificial membranes of phosphatidyl choline and microsomal phospholipid has almost identical secondary structure as does the soluble one. Phosphatidylcholines 63-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 7236798-5 1980 Cytochrome P-450 incorporated into the artificial membranes of phosphatidyl choline and microsomal phospholipid has almost identical secondary structure as does the soluble one. Phospholipids 99-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 7236798-6 1980 Data from circular dichroism suggest that the binding of the types I and II substrates to cytochrome P-450 incorporated into lecithin liposomes and microsomal lipid liposomes does not change the conformation of the polypeptide chain. Lecithins 125-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 6105054-0 1980 Correlations between cytochrome P-450 and oxidative metabolism of benzo[a]pyrene and 7-ethoxycoumarin in human liver in vitro and antipyrine elimination in vivo. Benzo(a)pyrene 66-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 7192358-6 1980 Addition of BHA to the medium inhibited growth of the culture on glucose and biosynthesis of cytochrome P-450 in these cells. benzohydroxamic acid 12-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 7192358-8 1980 Apparently, hydroxamic acids can affect the operation of the microsomal monooxigenase system containing cytochrome P-450. Hydroxamic Acids 12-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 6771263-2 1980 Two N,N-dimethylphentermine (N,N-dimethyl-2-amino-2-methyl-3-phenylpropane) substrates differing in deuterium substitution have been used to determine the intermolecular and intramolecular isotope effects associated with the cytochrome P-450-dependent N-demethylation of this substrate. n,n-dimethyl-2-amino-2-methyl-3-phenylpropane 29-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 225-241 6105054-0 1980 Correlations between cytochrome P-450 and oxidative metabolism of benzo[a]pyrene and 7-ethoxycoumarin in human liver in vitro and antipyrine elimination in vivo. 7-ethoxycoumarin 85-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 6105054-4 1980 The correlation of cytochrome P-450 content with aryl hydrocarbon hydroxylase activity in human liver biopsy samples was relatively good (r = 0.75, p < 0.001), but that with 7-ethoxycoumarin O-de-ethylase activity was much poorer (r = 0.42, p < 0.001). 7-ethoxycoumarin 177-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 6105054-6 1980 This poor correlation was caused mainly by the nonequal effects of polycyclic aromatic hydrocarbons on cytochrome P-450 and aryl hydrocarbon hydroxylase. Polycyclic Aromatic Hydrocarbons 67-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-152 7391974-0 1980 The characterization of an inhibitory complex formed with cytochrome P-450 and a metabolite of 1,1-disubstituted hydrazines. 1,1-disubstituted hydrazines 95-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 7379213-5 1980 The finding that singlet oxygen will oxidise a cell constituent into a powerful inducer is compatible with the hypothesis that excited states of oxygen and their oxidation products may play a central role in the induction of cytochrome P-450 and associated enzyme activities by many chemically unrelated inducers. Singlet Oxygen 17-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 225-241 7379213-5 1980 The finding that singlet oxygen will oxidise a cell constituent into a powerful inducer is compatible with the hypothesis that excited states of oxygen and their oxidation products may play a central role in the induction of cytochrome P-450 and associated enzyme activities by many chemically unrelated inducers. Oxygen 25-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 225-241 6772704-1 1980 The first step in ethanol metabolism is carried out by two enzyme systems: Alcohol dehydrogenase and cytochrome P-450. Ethanol 18-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-117 7391953-0 1980 Ligand interaction of sustituted pyridines with cytochrome P-450. Pyridines 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 7391953-1 1980 A series of pyridyl ketones and alkyl pyridines was evaluated as type II ligands for cytochrome P-450. pyridyl ketones 12-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 7391953-1 1980 A series of pyridyl ketones and alkyl pyridines was evaluated as type II ligands for cytochrome P-450. alkyl pyridines 32-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 7398652-0 1980 Visible and ultraviolet spectral transitions of camphor-bound cytochrome P-450. Camphor 48-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 7398652-2 1980 A systematic analysis of the low-spin to high-spin transition in the ultraviolet and visible absorption spectrum of camphor-bound cytochrome P-450 is presented. Camphor 116-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 7375915-2 1980 Antibody to the cytochrome P-450 induced by phenobarbital has relatively little or no effect on the aryl hydrocarbon hydroxylase activity of the same human cells. Phenobarbital 44-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 7381464-5 1980 This apparent magnetic anisotropy of the mercaptide complex is in the same direction, although smaller, than that observed for bacterial cytochrome P-450. mercaptide 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-153 7378257-2 1980 2 The apparent plasma clearance of propranolol was closely related both to the in vivo (antipyrine test) and in vitro (cytochrome P-450) indices of the activity of the hepatic mixed function oxidase system. Propranolol 35-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 7371884-2 1980 Spectral studies indicated that the inhibition was a consequence of danazol"s interfering with the functioning of mitochondrial cytochrome P-450, an essential component of the enzyme system involved in progesterone biosynthesis. Danazol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 7371884-2 1980 Spectral studies indicated that the inhibition was a consequence of danazol"s interfering with the functioning of mitochondrial cytochrome P-450, an essential component of the enzyme system involved in progesterone biosynthesis. Progesterone 202-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 7387676-0 1980 Protection by N-acetylcysteine of cyclophosphamide metabolism - related in vivo depression of mixed function oxygenase activity and in vitro denaturation of cytochrome P-450. Acetylcysteine 14-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 7387676-0 1980 Protection by N-acetylcysteine of cyclophosphamide metabolism - related in vivo depression of mixed function oxygenase activity and in vitro denaturation of cytochrome P-450. Cyclophosphamide 34-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 7373548-0 1980 Inhibition of in vitro cytochrome P-450-catalyzed reactions by substituted pyridines. Pyridines 75-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 7256184-4 1980 Serum triglyceride level was inversely related to the hepatic cytochrome P-450 content (r = 0.62, P less than 0.01). Triglycerides 6-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 6767272-1 1980 Cytochrome P-450-dependent monooxygenase systems, which metabolize endogenous as well as foriegn compounds, are found in hepatic and several extrahepatic tissues of mammals, including humans. foriegn compounds 89-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 7373548-1 1980 A series of substituted pyridines was investigated as inhibitors of cytochrome P-450-catalyzed reactions. Pyridines 24-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 6768169-0 1980 Induction of liver microsomal NADPH cytochrome C reductase and cytochrome P-450 by some new synthetic pyrethroids. Pyrethrins 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 6929480-0 1980 Chemical mechanisms for cytochrome P-450 hydroxylation: evidence for acylation of heme-bound dioxygen. Heme 82-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 6771479-0 1980 Induction of different species of cytochrome P-450 by coplanar and noncoplanar isomers of hexachlorobiphenyl. 2,2',3,4,5,6-Hexachlorobiphenyl 90-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 7388141-0 1980 [Induction of cytochrome P-450 and the immune response by free and covalently bound to albumin phenobarbital]. Phenobarbital 95-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 6929480-1 1980 Using isotopic tracer methods, we have shown that dihydrolipoic acid (2,3-thioctic acid) acylates the distal oxygen of ferrous oxygenated Pseudomonas cytochrome P-450, forming a transient acyl peroxide intermediate that facilitates oxygen-oxygen bond cleavage. acyl peroxide 188-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 6929480-1 1980 Using isotopic tracer methods, we have shown that dihydrolipoic acid (2,3-thioctic acid) acylates the distal oxygen of ferrous oxygenated Pseudomonas cytochrome P-450, forming a transient acyl peroxide intermediate that facilitates oxygen-oxygen bond cleavage. Oxygen 127-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 6929480-0 1980 Chemical mechanisms for cytochrome P-450 hydroxylation: evidence for acylation of heme-bound dioxygen. Oxygen 93-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 6929480-1 1980 Using isotopic tracer methods, we have shown that dihydrolipoic acid (2,3-thioctic acid) acylates the distal oxygen of ferrous oxygenated Pseudomonas cytochrome P-450, forming a transient acyl peroxide intermediate that facilitates oxygen-oxygen bond cleavage. Oxygen 127-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 6929480-1 1980 Using isotopic tracer methods, we have shown that dihydrolipoic acid (2,3-thioctic acid) acylates the distal oxygen of ferrous oxygenated Pseudomonas cytochrome P-450, forming a transient acyl peroxide intermediate that facilitates oxygen-oxygen bond cleavage. dihydrolipoic acid 50-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 6929480-1 1980 Using isotopic tracer methods, we have shown that dihydrolipoic acid (2,3-thioctic acid) acylates the distal oxygen of ferrous oxygenated Pseudomonas cytochrome P-450, forming a transient acyl peroxide intermediate that facilitates oxygen-oxygen bond cleavage. 2,3-thioctic acid 70-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 6929480-1 1980 Using isotopic tracer methods, we have shown that dihydrolipoic acid (2,3-thioctic acid) acylates the distal oxygen of ferrous oxygenated Pseudomonas cytochrome P-450, forming a transient acyl peroxide intermediate that facilitates oxygen-oxygen bond cleavage. Oxygen 109-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 7435083-0 1980 Characterization of the cytochrome P-450 species induced by trans-stilbene oxide. stilbene oxide 60-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 6775511-0 1980 On the possible relationship of cytochrome P-450 to alcohol metabolism: fundamental aspects of the microsomal hydroxylation system, including properties and interactions of the components. Alcohols 52-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 6775511-1 1980 Recent studies on the cytochrome P-450-containing enzyme system of liver microsomes have shown that the cytochrome is present as a number of distinct isozymes, one of which is induced in vivo by the administration of aromatic hydrocarbons, as well as several forms which are less well characterized and not known to be inducible. Hydrocarbons, Aromatic 217-238 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 6775511-6 1980 The possible effect of ethanol on these interactions should be considered in evaluating the reported inhibition by high concentrations of ethanol of reactions catalyzed by liver microsomal cytochrome P-450. Ethanol 23-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-205 6775511-6 1980 The possible effect of ethanol on these interactions should be considered in evaluating the reported inhibition by high concentrations of ethanol of reactions catalyzed by liver microsomal cytochrome P-450. Ethanol 138-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-205 7370330-1 1980 Thermal inactivation of cytochrome P-450 in different states (microsomes, highly purified and immobilized), characterized by the loss of catalytic activity in cumene hydroperoxide--dependent aniline hydroxylation has been studied in the temperature range 40-58 degrees C. The process of thermoinactivation is characterized by the first order rate constants. cumene hydroperoxide 159-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 6940467-4 1980 The loss of cytochrome P-450 elicited by IF-inducing agents is accompanied by a perturbation of heme metabolism associated with the dissociation of heme from cytochrome P-450. Heme 96-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 7370330-1 1980 Thermal inactivation of cytochrome P-450 in different states (microsomes, highly purified and immobilized), characterized by the loss of catalytic activity in cumene hydroperoxide--dependent aniline hydroxylation has been studied in the temperature range 40-58 degrees C. The process of thermoinactivation is characterized by the first order rate constants. aniline 191-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 7370330-2 1980 From the temperature dependence of these constants the energy of activation and the activation parameters of cytochrome P-450 thermoinactivation have been determined at 45 degrees C. In the presence of 20% glycerol the activation parameters of cytochrome P-450 thermoinactivation in microsomes in highly purified and immobilized states are very similar. Glycerol 206-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 7370330-2 1980 From the temperature dependence of these constants the energy of activation and the activation parameters of cytochrome P-450 thermoinactivation have been determined at 45 degrees C. In the presence of 20% glycerol the activation parameters of cytochrome P-450 thermoinactivation in microsomes in highly purified and immobilized states are very similar. Glycerol 206-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 244-260 7370331-0 1980 [Multiphasic character of the kinetics of cytochrome P-450 destruction in microsomal LM2- and LM4-forms in the reaction with cumene hydroperoxide]. cumene hydroperoxide 125-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 7370331-1 1980 Cytochrome P-450 destruction kinetics by cumene hydroperoxide has been studied in LM2 and LM4 microsomal and purified forms. cumene hydroperoxide 41-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 7370331-2 1980 Three destruction phases of cytochrome P-450 were shown to be observed irrespective of the source and integration degree, cytochrome P-450 pseudomonomolecular consumption rate constants being dependent in a complex way upon the cumene hydroperoxide initial concentration. cumene hydroperoxide 228-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 7370331-2 1980 Three destruction phases of cytochrome P-450 were shown to be observed irrespective of the source and integration degree, cytochrome P-450 pseudomonomolecular consumption rate constants being dependent in a complex way upon the cumene hydroperoxide initial concentration. cumene hydroperoxide 228-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 7370331-3 1980 The radical character of cytochrome P-450 destruction was proved by experiments with 1-naphtol. 1-naphthol 85-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 6086112-0 1980 [Effect of phospholipids on the activity of highly-purified liver microsome cytochrome P-450 in a reaction of aniline and naphthalene hydroxylation by hydroperoxides]. Phospholipids 11-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 6933961-0 1980 Ligand binding of safrole to cytochrome P-450. Safrole 18-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 6933961-1 1980 Safrole, a hepatocarcinogen, is converted by the microsomal mono-oxygenase system to a reactive intermediate which interacts with cytochrome P-450 to form a ligand complex. Safrole 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 6086112-0 1980 [Effect of phospholipids on the activity of highly-purified liver microsome cytochrome P-450 in a reaction of aniline and naphthalene hydroxylation by hydroperoxides]. aniline 110-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 6086112-0 1980 [Effect of phospholipids on the activity of highly-purified liver microsome cytochrome P-450 in a reaction of aniline and naphthalene hydroxylation by hydroperoxides]. naphthalene 122-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 6086112-0 1980 [Effect of phospholipids on the activity of highly-purified liver microsome cytochrome P-450 in a reaction of aniline and naphthalene hydroxylation by hydroperoxides]. Hydrogen Peroxide 151-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 6086112-1 1980 The effect of different phospholipids on the functional activity of highly purified cytochrome P-450 used as a co-substrate of cumene hydroperoxide was examined. Phospholipids 24-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 6086112-1 1980 The effect of different phospholipids on the functional activity of highly purified cytochrome P-450 used as a co-substrate of cumene hydroperoxide was examined. cumene hydroperoxide 127-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 7423530-2 1980 In contrast, several other sulfur containing compounds, tetraethyl thiuramidisulfide, diethyldithiocarbamic acid, thiourea or 1-methyl-2-mercaptoimidazole, which are inhibitors of non-cytochrome P-450 dependent amine oxidase systems, significantly prevented thioacetamide induced liver necrosis at 24 h. Notwithstanding, diphenhydramine, nicotinamide, trimethylamine and imipramine, which are substrates of this amino oxidase system, do not protect. Sulfur 27-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-200 6102422-0 1980 The role of cytochrome P-450-inducing agents in potentiaing the toxicity of fluroxene (2,2,2-trifluoroethyl vinyl ether). fluroxene 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 6102422-0 1980 The role of cytochrome P-450-inducing agents in potentiaing the toxicity of fluroxene (2,2,2-trifluoroethyl vinyl ether). fluroxene 87-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 7423530-2 1980 In contrast, several other sulfur containing compounds, tetraethyl thiuramidisulfide, diethyldithiocarbamic acid, thiourea or 1-methyl-2-mercaptoimidazole, which are inhibitors of non-cytochrome P-450 dependent amine oxidase systems, significantly prevented thioacetamide induced liver necrosis at 24 h. Notwithstanding, diphenhydramine, nicotinamide, trimethylamine and imipramine, which are substrates of this amino oxidase system, do not protect. tetraethyl thiuramidisulfide 56-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-200 7423530-2 1980 In contrast, several other sulfur containing compounds, tetraethyl thiuramidisulfide, diethyldithiocarbamic acid, thiourea or 1-methyl-2-mercaptoimidazole, which are inhibitors of non-cytochrome P-450 dependent amine oxidase systems, significantly prevented thioacetamide induced liver necrosis at 24 h. Notwithstanding, diphenhydramine, nicotinamide, trimethylamine and imipramine, which are substrates of this amino oxidase system, do not protect. Ditiocarb 86-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-200 7423530-2 1980 In contrast, several other sulfur containing compounds, tetraethyl thiuramidisulfide, diethyldithiocarbamic acid, thiourea or 1-methyl-2-mercaptoimidazole, which are inhibitors of non-cytochrome P-450 dependent amine oxidase systems, significantly prevented thioacetamide induced liver necrosis at 24 h. Notwithstanding, diphenhydramine, nicotinamide, trimethylamine and imipramine, which are substrates of this amino oxidase system, do not protect. 2-mercaptoimidazole 135-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-200 120229-0 1979 A model of the heme site of cytochrome P-450: characterization of a sulfhydryl- and imidazole-containing peptide-heme system in solution. Heme 15-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 115873-1 1979 A re-examination of the effects of copper chelates on the function of cytochrome P-450. Copper 35-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 115873-7 1979 In particular, it was found that the reported inhibition of cytochrome P-450-catalyzed hydroxylation reactions by copper-tyrosine is associated with an inhibition rather than a stimulation of the formation of hydrogen peroxide, the product of the dismutation of the superoxide radicals generated as a result of the decay of oxycytochrome P-450. copper-tyrosine 114-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 115873-7 1979 In particular, it was found that the reported inhibition of cytochrome P-450-catalyzed hydroxylation reactions by copper-tyrosine is associated with an inhibition rather than a stimulation of the formation of hydrogen peroxide, the product of the dismutation of the superoxide radicals generated as a result of the decay of oxycytochrome P-450. Hydrogen Peroxide 209-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 115873-7 1979 In particular, it was found that the reported inhibition of cytochrome P-450-catalyzed hydroxylation reactions by copper-tyrosine is associated with an inhibition rather than a stimulation of the formation of hydrogen peroxide, the product of the dismutation of the superoxide radicals generated as a result of the decay of oxycytochrome P-450. Superoxides 266-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 120229-0 1979 A model of the heme site of cytochrome P-450: characterization of a sulfhydryl- and imidazole-containing peptide-heme system in solution. sulfhydryl- and imidazole 68-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 120229-0 1979 A model of the heme site of cytochrome P-450: characterization of a sulfhydryl- and imidazole-containing peptide-heme system in solution. Peptides 105-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 120229-0 1979 A model of the heme site of cytochrome P-450: characterization of a sulfhydryl- and imidazole-containing peptide-heme system in solution. Heme 113-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 397836-2 1979 A special emphasis is laid upon the interaction of alcohols with terminal oxidase of the microsomal hydroxylating system, i.e. cytochrome P-450. Alcohols 51-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-143 41598-3 1979 Differences in the Tyr2-Cu2+ complex effects on the cumule hydroperoxide-dependent xenobiotics hydroxylation and lipid peroxidation catalyzed by various forms of cytochrome P-450, e. tyr2-cu2+ 19-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-181 526052-0 1979 Dinitrochlorobenzene: influence on the cytochrome P-450 system and mutagenic effects. Dinitrochlorobenzene 0-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 226536-0 1979 The effect of cytochrome P-450cam on the NMR relaxation rate of water protons. Water 64-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 226536-1 1979 Cytochrome P-450cam in the native, substrate-free state (Fe3+, S = 1/2) substantially reduces the NMR relaxation times, T1 and T2, of water protons. Water 134-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 40703-2 1979 With the exception of dioxolane, 4-methyldioxolane and 4-ethyldioxolane, these compounds interacted with ferric cytochrome P-450 to give complexes exhibiting type I optical difference spectra, and, after incubation with NADPH, spectra with peaks at about 430 nm. NADP 220-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 115595-3 1979 p-Aminophenol administration lowered the microsomal cytochrome P-450 and b5 content and decreased the activity of NADPH cytochrome c reductase in kidney, but not in liver. 4-aminophenol 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-74 41598-3 1979 Differences in the Tyr2-Cu2+ complex effects on the cumule hydroperoxide-dependent xenobiotics hydroxylation and lipid peroxidation catalyzed by various forms of cytochrome P-450, e. Hydrogen Peroxide 59-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-181 41598-5 1979 The data obtained suggest that the efficiency of the inhibitory effect of the Tyr2-Cu2+ complex depends on the type of cosubstrates (NADPH, cumole hydroperoxide) and substrates used as well as on the form of cytochrome P-450. tyr2-cu2+ 78-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 208-224 41598-5 1979 The data obtained suggest that the efficiency of the inhibitory effect of the Tyr2-Cu2+ complex depends on the type of cosubstrates (NADPH, cumole hydroperoxide) and substrates used as well as on the form of cytochrome P-450. NADP 133-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 208-224 41598-5 1979 The data obtained suggest that the efficiency of the inhibitory effect of the Tyr2-Cu2+ complex depends on the type of cosubstrates (NADPH, cumole hydroperoxide) and substrates used as well as on the form of cytochrome P-450. cumole hydroperoxide 140-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 208-224 116333-1 1979 Delta 9-THC and 11-OH-delta 9-THC appear to in vivo affect the metabolism of both Type I and Type II substrates involving cytochrome P-450 as well as to affect the metabolism of a substrate requiring cytochrome P-448. Dronabinol 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 43918-0 1979 The apparent loss of cytochrome P-450 associated with metabolic activation of carbon tetrachloride. Carbon Tetrachloride 78-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 43918-3 1979 The addition of methylviologen also enhanced the carbon tetrachloride-induced loss of cytochrome P-450, while the apparent content of cytochrome b5 and the activity of NADPH-cytochrome c reductase remained unchanged. Carbon Tetrachloride 49-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 43918-4 1979 Under a strong inhibition of lipid peroxidation by addition of EDTA, carbon tetrachloride induced a clear loss of cytochrome P-450 to the extent similar to that seen in the absence of EDTA. Edetic Acid 63-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 43918-4 1979 Under a strong inhibition of lipid peroxidation by addition of EDTA, carbon tetrachloride induced a clear loss of cytochrome P-450 to the extent similar to that seen in the absence of EDTA. Carbon Tetrachloride 69-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 43918-5 1979 These results indicate that cytochrome P-450 is directly degraded in association with the reductive metabolism of carbon tetrachloride by cytochrome P-450. Carbon Tetrachloride 114-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 43918-5 1979 These results indicate that cytochrome P-450 is directly degraded in association with the reductive metabolism of carbon tetrachloride by cytochrome P-450. Carbon Tetrachloride 114-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-154 519812-0 1979 A model system of cytochrome P-450: hydroxylation of aniline by iron- or hemin-thiol compound systems. aniline 53-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 519812-0 1979 A model system of cytochrome P-450: hydroxylation of aniline by iron- or hemin-thiol compound systems. Iron 64-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 519812-0 1979 A model system of cytochrome P-450: hydroxylation of aniline by iron- or hemin-thiol compound systems. hemin-thiol 73-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 116333-1 1979 Delta 9-THC and 11-OH-delta 9-THC appear to in vivo affect the metabolism of both Type I and Type II substrates involving cytochrome P-450 as well as to affect the metabolism of a substrate requiring cytochrome P-448. 11-oh-delta 9-thc 16-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 507813-0 1979 Induction by manganese, ethanol, phenobarbital, and herbicides of microsomal cytochrome P-450 in higher plant tissues. Manganese 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 507813-0 1979 Induction by manganese, ethanol, phenobarbital, and herbicides of microsomal cytochrome P-450 in higher plant tissues. Ethanol 24-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 507813-0 1979 Induction by manganese, ethanol, phenobarbital, and herbicides of microsomal cytochrome P-450 in higher plant tissues. Phenobarbital 33-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 383076-0 1979 The role of cytochrome P-450 forms in 2-aminoanthracene and benz[alpha]pyrene mutagenesis. 2-anthramine 38-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 455885-3 1979 Plasma antipyrine clearance and cytochrome P-450 content in biopsies were related to propranolol elimination from plasma, the best fit being obtained with the clearance values. Propranolol 85-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 383076-0 1979 The role of cytochrome P-450 forms in 2-aminoanthracene and benz[alpha]pyrene mutagenesis. benz[alpha]pyrene 60-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 109441-1 1979 The interaction between cytochrome P-450 and NADPH-cytochrome c reductase during catalysis has been investigated with a reconstituted monooxygenase system composed of the two purified enzyme components and synthetic phospholipid. Phospholipids 216-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 109441-4 1979 In agreement with this equation, the observed Vmax for benzphetamine N-demethylation was found to be directly proportional to the calculated concentration of the cytochrome P-450 . Benzphetamine 55-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-178 109441-4 1979 In agreement with this equation, the observed Vmax for benzphetamine N-demethylation was found to be directly proportional to the calculated concentration of the cytochrome P-450 . Nitrogen 69-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-178 456582-0 1979 Subzero temperature studies of microsomal cytochrome P-450: O-dealkylation of 7-ethoxycoumarin coupled to single turnover. 7-ethoxycoumarin 78-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 447633-0 1979 The metabolism of benzo(alpha)pyrene by cytochrome P-450 in transformable and nontransformable C3H mouse fibroblasts. Benzo[alpha]pyrene 18-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 762169-0 1979 Studies on the destruction of adrenal and testicular cytochrome P-450 by spironolactone. Spironolactone 73-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 221013-3 1979 The results show that the heme plane of cytochrome P-450 lies in the same plane as the membrane surface, whereas the cytochrome b5 heme plane has a random orientation. Heme 26-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 483858-9 1979 Any treatment of microsomes which resulted in a reduction of cytochrome P-450 also produced a concomitant fall in N-oxidation of the pyridines. Pyridines 133-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 428008-6 1979 Metyrapone and SKF 525-A inhibit covalent binding of the hormone to cytoplasmic macromolecules, which suggests participation of the cytochrome P-450 system in covalent binding of the hormone. Metyrapone 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 223025-1 1979 Cytochrome P450 in the mitochondria of the adrenal cortex functions in the monooxygenation reactions for the biosynthesis of various steroid hormones, such as cholesterol side chain cleavage, hydroxylation at 11 beta-position and that at 18-position of the steroid structure. Steroids 133-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 223025-1 1979 Cytochrome P450 in the mitochondria of the adrenal cortex functions in the monooxygenation reactions for the biosynthesis of various steroid hormones, such as cholesterol side chain cleavage, hydroxylation at 11 beta-position and that at 18-position of the steroid structure. Cholesterol 159-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 223025-1 1979 Cytochrome P450 in the mitochondria of the adrenal cortex functions in the monooxygenation reactions for the biosynthesis of various steroid hormones, such as cholesterol side chain cleavage, hydroxylation at 11 beta-position and that at 18-position of the steroid structure. Steroids 257-264 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 223025-7 1979 These effects are important from the point of view that the primary role of the heme of cytochrome P450 is the activation of molecular oxygen. Heme 80-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 223025-7 1979 These effects are important from the point of view that the primary role of the heme of cytochrome P450 is the activation of molecular oxygen. Oxygen 135-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 112378-0 1979 Inhibition of hepatic microsomal cytochrome P-450-dependent monooxygenase reactions by fatty acyl CoA. fatty 87-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 112378-0 1979 Inhibition of hepatic microsomal cytochrome P-450-dependent monooxygenase reactions by fatty acyl CoA. Coenzyme A 98-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 428008-6 1979 Metyrapone and SKF 525-A inhibit covalent binding of the hormone to cytoplasmic macromolecules, which suggests participation of the cytochrome P-450 system in covalent binding of the hormone. Proadifen 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 35794-0 1979 Iopronic acid interaction with hepatic microsomal cytochrome P-450: relationship between chemical structure and binding. iopronic acid 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 284396-0 1979 Self-catalyzed destruction of cytochrome P-450: covalent binding of ethynyl sterols to prosthetic heme. ethynyl sterols 68-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 284396-0 1979 Self-catalyzed destruction of cytochrome P-450: covalent binding of ethynyl sterols to prosthetic heme. Heme 98-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 284396-1 1979 The hepatic pigment accumulated as a consequence of the self-catalyzed destruction of cytochrome P-450 by norethisterone (17-hydroxy-19-nor-17 alpha-pregn-4-en-20-yn-3-one), after acidic methylation and purification, consists of two virtually identical, probably isomeric, porphyrins. Norethindrone 106-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 284396-1 1979 The hepatic pigment accumulated as a consequence of the self-catalyzed destruction of cytochrome P-450 by norethisterone (17-hydroxy-19-nor-17 alpha-pregn-4-en-20-yn-3-one), after acidic methylation and purification, consists of two virtually identical, probably isomeric, porphyrins. Norethindrone 122-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 284396-1 1979 The hepatic pigment accumulated as a consequence of the self-catalyzed destruction of cytochrome P-450 by norethisterone (17-hydroxy-19-nor-17 alpha-pregn-4-en-20-yn-3-one), after acidic methylation and purification, consists of two virtually identical, probably isomeric, porphyrins. Porphyrins 273-283 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 284396-4 1979 Cytochrome P-450 is therefore destroyed by self-catalyzed addition of norethisterone to its heme prosthetic group. Norethindrone 70-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 284396-4 1979 Cytochrome P-450 is therefore destroyed by self-catalyzed addition of norethisterone to its heme prosthetic group. Heme 92-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 284396-5 1979 Cytochrome P-450 is also destroyed by norgestrel (13-ethyl-17-hydroxyl-18, 19-dinor-17 alpha-pregn-4-en-20-yn-3-one) and by 1-ethynylcyclohexanol but not by 17-hydroxy-19-nor-17alpha-pregn-4,20-dien-3-one. Norgestrel 38-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 284396-5 1979 Cytochrome P-450 is also destroyed by norgestrel (13-ethyl-17-hydroxyl-18, 19-dinor-17 alpha-pregn-4-en-20-yn-3-one) and by 1-ethynylcyclohexanol but not by 17-hydroxy-19-nor-17alpha-pregn-4,20-dien-3-one. 13-ethyl-17-hydroxyl-18, 19-dinor-17 alpha-pregn-4-en-20-yn-3-one 50-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 284396-5 1979 Cytochrome P-450 is also destroyed by norgestrel (13-ethyl-17-hydroxyl-18, 19-dinor-17 alpha-pregn-4-en-20-yn-3-one) and by 1-ethynylcyclohexanol but not by 17-hydroxy-19-nor-17alpha-pregn-4,20-dien-3-one. 1-Ethynyl-1-cyclohexanol 124-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 284396-5 1979 Cytochrome P-450 is also destroyed by norgestrel (13-ethyl-17-hydroxyl-18, 19-dinor-17 alpha-pregn-4-en-20-yn-3-one) and by 1-ethynylcyclohexanol but not by 17-hydroxy-19-nor-17alpha-pregn-4,20-dien-3-one. 17-hydroxy-19-nor-17alpha-pregn-4,20-dien-3-one 157-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 42248-0 1979 NADPH reduction of cytochrome P-450 at different integrational levels of the enzyme system. NADP 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 42248-2 1979 The aerobic NADPH reduction of microsomes, solubilized and reconstituted systems follows a biphasic kinetics, the two phases are attributed to associated state (cluster) and random cytochrome P-450 reduction. NADP 12-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 42252-0 1979 Electrochemical investigations on the oxygen activation by cytochrome P-450. Oxygen 38-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 42252-7 1979 In contrast the cytochrome P-450 catalyzed NADPH-dependent reaction with the same substrate gives corticosterone, O2- represents only an intermediate in the activation of oxygen and is not the "activated oxygen" species. NADP 43-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 42252-7 1979 In contrast the cytochrome P-450 catalyzed NADPH-dependent reaction with the same substrate gives corticosterone, O2- represents only an intermediate in the activation of oxygen and is not the "activated oxygen" species. Corticosterone 98-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 42252-7 1979 In contrast the cytochrome P-450 catalyzed NADPH-dependent reaction with the same substrate gives corticosterone, O2- represents only an intermediate in the activation of oxygen and is not the "activated oxygen" species. Oxygen 114-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 42252-7 1979 In contrast the cytochrome P-450 catalyzed NADPH-dependent reaction with the same substrate gives corticosterone, O2- represents only an intermediate in the activation of oxygen and is not the "activated oxygen" species. Oxygen 171-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 42252-9 1979 The interaction of adsorbed cytochrome P-450 on the electrode surface with the reduced oxygen species in the absence of NADPH was studied. Oxygen 87-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 42252-12 1979 In a model of electro-enzyme-reactor several substrates were hydroxylated by microsomal cytochrome P-450 with cathodically reduced oxygen which substitutes NADPH. Oxygen 131-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 42252-12 1979 In a model of electro-enzyme-reactor several substrates were hydroxylated by microsomal cytochrome P-450 with cathodically reduced oxygen which substitutes NADPH. NADP 156-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 117659-1 1979 Cytochrome P-450 and NADPH-cytochrome P-450 reductase were covalently attached to Sepharose 4B in different ways in order to find out factors which are important for the organization of the individual components to a catalytically active system. Sepharose 82-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 229672-3 1979 The present study of the kinetics of reduction of cytochrome P-450cam by reduced putidaredoxin has shown that the reaction obeys first order kinetics with a rate constant of 33 s-1 at 25 degrees C with respect to: 1) the appearance of the carbon monoxide complex of Fe(II) cytochrome P-450cam; 2) the disappearance of the 645 nm absorbance band of high-spin Fe(III) cytochrome P-450cam; and 3) the disappearance of the g = 1.94 EPR signal of reduced putidaredoxin. Carbon Monoxide 239-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 229673-1 1979 Spin state transitions of membrane-bound cytochrome P-450 were investigated by difference spectrophotometry using the "D"-charge transfer absorbance band at 645 nm as a measure of the amount of hemin iron present in the 5-coordinated state. Iron 200-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 229673-2 1979 The magnitude of the "D"-absorbance band in the absence of exogenous substrates, e.g., the concentration of native high spin cytochrome P-450, was evaluated from the difference in absorbance at 645 nm between ferric cytochrome P-450 and the carbon monoxide derivative of the pigment in its ferrous state. Carbon Monoxide 241-256 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 229673-3 1979 The contribution of the native high spin species to the total cytochrome P-450 content of microsomes was calculated to be between 40% and 65% after induction with phenobarbital and polycyclic hydrocarbons, respectively. Phenobarbital 163-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 517003-4 1979 Liposome-bound cytochrome P-450 has a higher dimethylaniline, aniline and p-nitroanisole hydroxylase activity than its soluble form. N,N-dimethylaniline 45-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 229673-3 1979 The contribution of the native high spin species to the total cytochrome P-450 content of microsomes was calculated to be between 40% and 65% after induction with phenobarbital and polycyclic hydrocarbons, respectively. Hydrocarbons, Cyclic 181-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 229673-5 1979 Further, the steady state concentrations of high spin cytochrome P-450, observed in the presence of reduced pyridine nucleotides, suggest that the rate limiting step for microsomal mixed function oxidation reactions is variable and dependent on the substrate under investigation. pyridine nucleotides 108-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 229680-1 1979 Studies on model complexes have supported the presence of a mercaptide as the fifth ligand of cytochrome P-450 monooxygenases. mercaptide 60-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 229680-2 1979 When alcohol or thiol ligands are added to the sixth coordination position of a five-coordinated 4-nitrobenzene thiolate complex of FeIII protoporphyrin IX dimethyl ester chloride low spin complexes with optical and EPR-spectra very similar to cytochrome P-450 are obtained. Alcohols 5-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 244-260 229680-2 1979 When alcohol or thiol ligands are added to the sixth coordination position of a five-coordinated 4-nitrobenzene thiolate complex of FeIII protoporphyrin IX dimethyl ester chloride low spin complexes with optical and EPR-spectra very similar to cytochrome P-450 are obtained. Sulfhydryl Compounds 16-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 244-260 229680-2 1979 When alcohol or thiol ligands are added to the sixth coordination position of a five-coordinated 4-nitrobenzene thiolate complex of FeIII protoporphyrin IX dimethyl ester chloride low spin complexes with optical and EPR-spectra very similar to cytochrome P-450 are obtained. 4-nitrobenzene thiolate 97-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 244-260 229680-2 1979 When alcohol or thiol ligands are added to the sixth coordination position of a five-coordinated 4-nitrobenzene thiolate complex of FeIII protoporphyrin IX dimethyl ester chloride low spin complexes with optical and EPR-spectra very similar to cytochrome P-450 are obtained. ferric sulfate 132-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 244-260 229680-2 1979 When alcohol or thiol ligands are added to the sixth coordination position of a five-coordinated 4-nitrobenzene thiolate complex of FeIII protoporphyrin IX dimethyl ester chloride low spin complexes with optical and EPR-spectra very similar to cytochrome P-450 are obtained. protoporphyrin ix dimethyl ester chloride 138-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 244-260 229682-7 1979 Measurements of the reduction of succinylated cytochrome c using purified cytochrome P-450 and the flavoprotein, NADPH-cytochrome P-450 reductase, directly demonstrate the formation of superoxide anions. Superoxides 185-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-145 517003-4 1979 Liposome-bound cytochrome P-450 has a higher dimethylaniline, aniline and p-nitroanisole hydroxylase activity than its soluble form. aniline 53-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 517003-5 1979 Only the aniline hydroxylase activity of microsomal, proteoliposomal and soluble cytochrome P-450 was inhibited by tyrosine . Tyrosine 115-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 517003-7 1979 The inhibitory action of the Tyr2-Cu2+ complex on the other hydroxylase activities depended on the cytochrome P-450 forms and the type of cosubstrates and substrates used. tyr2-cu2+ 29-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 517006-0 1979 Mercaptide chelated protoheme: a model compound for cytochrome P-450. mercaptide 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 517006-1 1979 A mercaptide chelated heme having the spectral characteristics of cytochrome P-450 has been prepared by coupling di-3-amino-propyl-disulfide to protoheme, followed by reduction and disulfide cleavage with sodium dithionithe. mercaptide 2-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 517006-1 1979 A mercaptide chelated heme having the spectral characteristics of cytochrome P-450 has been prepared by coupling di-3-amino-propyl-disulfide to protoheme, followed by reduction and disulfide cleavage with sodium dithionithe. Heme 22-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 517006-1 1979 A mercaptide chelated heme having the spectral characteristics of cytochrome P-450 has been prepared by coupling di-3-amino-propyl-disulfide to protoheme, followed by reduction and disulfide cleavage with sodium dithionithe. di-3-amino-propyl-disulfide 113-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 517006-1 1979 A mercaptide chelated heme having the spectral characteristics of cytochrome P-450 has been prepared by coupling di-3-amino-propyl-disulfide to protoheme, followed by reduction and disulfide cleavage with sodium dithionithe. Heme 144-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 517006-1 1979 A mercaptide chelated heme having the spectral characteristics of cytochrome P-450 has been prepared by coupling di-3-amino-propyl-disulfide to protoheme, followed by reduction and disulfide cleavage with sodium dithionithe. Disulfides 131-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 517006-1 1979 A mercaptide chelated heme having the spectral characteristics of cytochrome P-450 has been prepared by coupling di-3-amino-propyl-disulfide to protoheme, followed by reduction and disulfide cleavage with sodium dithionithe. sodium dithionithe 205-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 517008-0 1979 Quantum chemical interpretation of the spectral properties of the CO and O2 complexes of hemoglobin and cytochrome P-450. Carbon Monoxide 66-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 517008-0 1979 Quantum chemical interpretation of the spectral properties of the CO and O2 complexes of hemoglobin and cytochrome P-450. Oxygen 73-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 517008-1 1979 The electronic transitions of CO and O2 complexes of hemoglobin and cytochrome P-450 were calculated using a PPP method extended for metal complexes. Carbon Monoxide 30-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 517008-1 1979 The electronic transitions of CO and O2 complexes of hemoglobin and cytochrome P-450 were calculated using a PPP method extended for metal complexes. Oxygen 37-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 517008-1 1979 The electronic transitions of CO and O2 complexes of hemoglobin and cytochrome P-450 were calculated using a PPP method extended for metal complexes. Metals 133-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 517008-2 1979 The calculations show that the unusual spectral properties of cytochrome P-450 are very sensitive to the iron-sulfur bond distance. Iron 105-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 517008-2 1979 The calculations show that the unusual spectral properties of cytochrome P-450 are very sensitive to the iron-sulfur bond distance. Sulfur 110-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 517008-3 1979 It is suggested from these calculations that for the conversion of cytochrome P-450 to cytochrome P-420 an increase of the iron-sulfur bond distance of only about 0.2 A is sufficient. Iron 123-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 517008-3 1979 It is suggested from these calculations that for the conversion of cytochrome P-450 to cytochrome P-420 an increase of the iron-sulfur bond distance of only about 0.2 A is sufficient. Sulfur 128-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 517008-4 1979 The anomalous Soret band of the CO complex as well as the normal Soret band of the O2 complex of cytochrome P-450 are explicable assuming a mercaptide sulfur as fifth ligand. mercaptide sulfur 140-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 517010-3 1979 The two maxima of the nonequilibrium form of cytochrome P-450 without substrate in the visible absorption spectrum (alpha-band, beta-band) and the ratio of their intensities indicate the low-spin character of the heme iron. Heme 213-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 517010-3 1979 The two maxima of the nonequilibrium form of cytochrome P-450 without substrate in the visible absorption spectrum (alpha-band, beta-band) and the ratio of their intensities indicate the low-spin character of the heme iron. Iron 218-222 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 517010-4 1979 These spectral properties give evidence for a reduced cytochrome P-450 with two heme-linked axial ligands. Heme 80-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 31893-0 1979 Cumene hydroperoxide-supported microsomal hydroxylations of warfarin--a probe of cytochrome P-450 multiplicity and specificity. cumene hydroperoxide 0-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 31893-0 1979 Cumene hydroperoxide-supported microsomal hydroxylations of warfarin--a probe of cytochrome P-450 multiplicity and specificity. Warfarin 60-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 119555-0 1979 [Cytochrome P-450 and transformation from 18-hydroxycorticosterone to aldosterone]. Aldosterone 70-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 1-17 552352-8 1979 Ethanol inhibition of drug metabolism in vitro appears to result from a modification of the lipophilic milieu that surrounds the cytochrome P-450 in the microsomal membrane. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 441518-4 1979 However, single doses of 400 mg/kg and accumulative doses of 500 mg/kg do cause a decrease in cytochrome P450 content as part of the dantrolene induced inhibition of the hepatic MFO system. Dantrolene 133-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-109 118858-7 1979 The interface between plants and insects is enormous, and therefore it appears probable that cytochrome P-450 will one day be shown to be important in the conversion/utilization of other plant secondary substances, e.g., synthesis of cholesterol from phytosterols, and sex pheromones from olefins, terpenes, and alkaloids. Cholesterol 234-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 118858-7 1979 The interface between plants and insects is enormous, and therefore it appears probable that cytochrome P-450 will one day be shown to be important in the conversion/utilization of other plant secondary substances, e.g., synthesis of cholesterol from phytosterols, and sex pheromones from olefins, terpenes, and alkaloids. Phytosterols 251-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 118858-7 1979 The interface between plants and insects is enormous, and therefore it appears probable that cytochrome P-450 will one day be shown to be important in the conversion/utilization of other plant secondary substances, e.g., synthesis of cholesterol from phytosterols, and sex pheromones from olefins, terpenes, and alkaloids. Alkenes 289-296 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 118858-7 1979 The interface between plants and insects is enormous, and therefore it appears probable that cytochrome P-450 will one day be shown to be important in the conversion/utilization of other plant secondary substances, e.g., synthesis of cholesterol from phytosterols, and sex pheromones from olefins, terpenes, and alkaloids. Terpenes 298-306 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 118858-7 1979 The interface between plants and insects is enormous, and therefore it appears probable that cytochrome P-450 will one day be shown to be important in the conversion/utilization of other plant secondary substances, e.g., synthesis of cholesterol from phytosterols, and sex pheromones from olefins, terpenes, and alkaloids. Alkaloids 312-321 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 441518-4 1979 However, single doses of 400 mg/kg and accumulative doses of 500 mg/kg do cause a decrease in cytochrome P450 content as part of the dantrolene induced inhibition of the hepatic MFO system. mfo 178-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-109 32885-0 1978 The degradation of different forms of cytochrome P-450 in vivo by fluroxene and allyl-iso-propylacetamide. fluroxene 66-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 217389-0 1978 Trophoblast estrogen synthetase stimulation by dibutyryl cyclic AMP and theophylline: increase in cytochrome P-450 content. Bucladesine 47-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 32885-0 1978 The degradation of different forms of cytochrome P-450 in vivo by fluroxene and allyl-iso-propylacetamide. Allylisopropylacetamide 80-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 711851-5 1978 AHH, a cytochrome P-450 dependent carcinogen-metabolizing enzyme appears to play an important role in the activation of polycyclic hydrocarbons into reactive moieties that can bind to DNA and that may directly induce cancer. Hydrocarbons, Cyclic 120-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 719999-4 1978 Hexachlorobenzene administration for 45 days resulted in the development of porphyria in rats, which biochemically closely resembles symptomatic porphyria in humans, with elevation of urinary uroporphyrin excretion, hepatic uroporphyrin content, and hepatic cytochrome P-450 content, in addition to appearance of porphyrins of the isocoproporphyrin (P1) series in the faeces. Hexachlorobenzene 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-274 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Aminopyrine 56-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-262 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Aminopyrine 56-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 339-355 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Benzo(a)pyrene 88-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-262 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Benzo(a)pyrene 88-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 339-355 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Hexachlorobenzene 210-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-262 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Hexachlorobenzene 210-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 339-355 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Hexachlorobenzene 303-320 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-262 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Hexachlorobenzene 303-320 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 339-355 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Phenobarbital 430-443 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-262 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Phenobarbital 430-443 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 339-355 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Methylcholanthrene 447-467 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-262 719999-8 1978 Study in vitro of the kinetics of two reactions, namely aminopyrine N-demethylation and 3,4-benzpyrene hydroxylation, catalysed by the hepatic microsomal cytochrom P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450-dependent enzyme system, suggested that hexachlorobenzene induced a form of cytochrome P-450 with different catalytic properties from those of forms induced by either phenobarbital or 3-methylcholanthrene. Methylcholanthrene 447-467 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 339-355 722999-0 1978 Glutathione depletion by aniline analogs in vitro associated with liver microsomal cytochrome P-450. Glutathione 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 722999-0 1978 Glutathione depletion by aniline analogs in vitro associated with liver microsomal cytochrome P-450. aniline 25-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 722999-1 1978 Enzymic depletion of glutathione (GSH) in vitro by aniline analogs was mostly dependent on the cytochrome P-450 level in liver microsomes. Glutathione 34-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 722999-1 1978 Enzymic depletion of glutathione (GSH) in vitro by aniline analogs was mostly dependent on the cytochrome P-450 level in liver microsomes. Glutathione 21-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 722999-1 1978 Enzymic depletion of glutathione (GSH) in vitro by aniline analogs was mostly dependent on the cytochrome P-450 level in liver microsomes. aniline 51-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 17539139-7 1978 Studies of mixed function oxidases and cytochrome P-450 system in male gonads demonstrated that the presence of AHH, EH, and GSH-ST implicate activation and detoxification of polycyclic hydrocarbons. Glutathione 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 28473-0 1978 Reduction of tertiary amine N-oxides by cytochrome P-450. tertiary amine 13-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 28473-0 1978 Reduction of tertiary amine N-oxides by cytochrome P-450. n-oxides 28-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 28181-0 1978 Complexes of trivalent oxygenated phosphorus compounds with cytochrome P-450 and cytochrome P-420: the origin of double Soret spectra. Phosphorus 34-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 17539139-7 1978 Studies of mixed function oxidases and cytochrome P-450 system in male gonads demonstrated that the presence of AHH, EH, and GSH-ST implicate activation and detoxification of polycyclic hydrocarbons. Hydrocarbons, Cyclic 175-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 206545-0 1978 Studies of the oxygen binding site of cytochrome P-450. Oxygen 15-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 208787-0 1978 A model of cytochrome P-450; optical and EPR properties of a thiol-containing peptide-hemin system and its activity of aniline hydroxylation. Sulfhydryl Compounds 61-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 208787-0 1978 A model of cytochrome P-450; optical and EPR properties of a thiol-containing peptide-hemin system and its activity of aniline hydroxylation. Peptides 78-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 208787-0 1978 A model of cytochrome P-450; optical and EPR properties of a thiol-containing peptide-hemin system and its activity of aniline hydroxylation. Hemin 86-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 208787-0 1978 A model of cytochrome P-450; optical and EPR properties of a thiol-containing peptide-hemin system and its activity of aniline hydroxylation. aniline 119-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 647071-0 1978 [Kinetic and spectral parameters of the amines oxidative N-dealkylation with participation of the liver microsomal cytochrome P-450. Amines 40-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 96251-1 1978 Treatment of animals with cobaltous chloride caused decreases in hepatic, pulmonary and renal cytochrome P-450, and alterations in levels of other components of microsomal mixed-function oxidases, which can alter the rate of biotransformation of certain drug substrates. cobaltous chloride 26-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 647071-2 1978 Kinetics of demethylation of a number of amines involving hepatic microsomal cytochrome P-450 and organic hydroperoxides (tret-butyl- and cumylhydroperoxide) have been investigated. Amines 41-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 647071-3 1978 Decomposition rate constants for the substrate-cytochrome P-450-ROOH complexes have been determined in a generalized form. Lipid Peroxides 64-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 23870-0 1978 [Kinetics of N-dealkylation of amines with participation of microsomal cytochrome P-450]. Nitrogen 13-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 647840-0 1978 The affinity of cytochrome P-450 for ethyl isocyanide: an explanation of double soret spectra. ethyl isocyanide 37-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 23870-0 1978 [Kinetics of N-dealkylation of amines with participation of microsomal cytochrome P-450]. Amines 31-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 23870-1 1978 Within the temperature range of 20-37 degrees C the kinetics of the demethylation reactions of a variety of amines with participation of hepatic microsomal cytochrome P-450, NADPH and O2 has been studied. Amines 108-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 23870-6 1978 The nature of the limiting step of the enzymatic amine demethylation involving cytochrome P-450 of hepatic microsomes is discussed. Amines 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 318378-0 1978 Induction of multiple cytochrome P-450 species in housefly microsomes--SDS-gel electrophoresis studies. Sodium Dodecyl Sulfate 71-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 318378-2 1978 Microsomal fractions isolated from various housefly strains have been characterized with respect to multiple forms of cytochrome P-450 by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Sodium Dodecyl Sulfate 138-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 318378-2 1978 Microsomal fractions isolated from various housefly strains have been characterized with respect to multiple forms of cytochrome P-450 by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. polyacrylamide 161-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 318378-10 1978 Induction with alpha-pinene and phenobarbital was expressed by a shift of the maximum absorbance at 452 nm in the CO-difference spectrum to lower wavelengths in the NAIDM strain; whereas, beta-naphthoflavone, although increasing the amount of cytochrome P-450, did not change the wavelength of maximum absorbance. alpha-pinene 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-259 318378-10 1978 Induction with alpha-pinene and phenobarbital was expressed by a shift of the maximum absorbance at 452 nm in the CO-difference spectrum to lower wavelengths in the NAIDM strain; whereas, beta-naphthoflavone, although increasing the amount of cytochrome P-450, did not change the wavelength of maximum absorbance. Phenobarbital 32-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-259 318378-10 1978 Induction with alpha-pinene and phenobarbital was expressed by a shift of the maximum absorbance at 452 nm in the CO-difference spectrum to lower wavelengths in the NAIDM strain; whereas, beta-naphthoflavone, although increasing the amount of cytochrome P-450, did not change the wavelength of maximum absorbance. naidm 165-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-259 212769-0 1978 Spectral evidence for weak ligand in sixth position of hepatic microsomal cytochrome P-450 low spin ferric iron in vivo. ferric sulfate 100-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 25763-3 1977 The fluroxene molecule (2,2,2-trifluroethyl vinyl ether) is composed of two moieties; both are toxic as a result of their metabolism: the vinyl moiety destroys heme of cytochrome P-450 while being metabolized to the final product, CO2. fluroxene 4-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-184 25763-3 1977 The fluroxene molecule (2,2,2-trifluroethyl vinyl ether) is composed of two moieties; both are toxic as a result of their metabolism: the vinyl moiety destroys heme of cytochrome P-450 while being metabolized to the final product, CO2. 2,2,2-trifluroethyl vinyl ether 24-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-184 25763-3 1977 The fluroxene molecule (2,2,2-trifluroethyl vinyl ether) is composed of two moieties; both are toxic as a result of their metabolism: the vinyl moiety destroys heme of cytochrome P-450 while being metabolized to the final product, CO2. Heme 160-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-184 25763-7 1977 Fluroxene metabolism and toxicity are modified by drugs metabolized by or affecting the activity of the microsomal cytochrome P-450-system or enzymes involved in ethanol metabolism. fluroxene 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 25763-7 1977 Fluroxene metabolism and toxicity are modified by drugs metabolized by or affecting the activity of the microsomal cytochrome P-450-system or enzymes involved in ethanol metabolism. Ethanol 162-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 925482-5 1977 This was confirmed by studies of the effect of ethinyl estradiol on the level of microsomal cytochrome P-450 over a 10-day period. Ethinyl Estradiol 47-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 925482-2 1977 After 5 days of ethinyl estradiol, 5 mg/kg/day, there was a significant decrease in the activity of ethylmorphine-N-demethylase and in cytochrome P-450, cytochrome b2, and NADPH cytochrome c reductase. Ethinyl Estradiol 16-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 334441-0 1977 Regulation of cytochrome P-450-dependent microsomal drug-metabolizing enzymes by nickel, cobalt, and iron. Nickel 81-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 925482-4 1977 After 5 days of ethinyl estradiol administration, the rate of degradation of cytochrome P-450 apoprotein was reduced (half-life of 50 hr compared to 24 hr in control), and their relative rate of synthesis was likewise reduced, indicating that a new steady state of protein turnover associated with reduced synthesis rate had been reached. Ethinyl Estradiol 16-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 611643-0 1977 The additive effects of progestins on testosterone-stimulated hepatic ethylmorphine metabolism and cytochrome P-450 content. Testosterone 38-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 202259-0 1977 Relationship between the reduction of oxygen, artificial acceptors and cytochrome P-450 by NADPH--cytochrome c reductase. Oxygen 38-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 202259-5 1977 In contrast with the process of electron transfer to cytochrome c, the rate of reduction of cytochrome P-450 and the rate of oxidation of adrenaline in liver microsomal fraction are correlated. Epinephrine 138-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 588632-0 1977 [O-demethylation kinetics of anisole and p-chloroanisole by cumene hydroperoxide and cytochrome P-450 of liver microsomes]. 4-chloroanisole 41-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 334441-0 1977 Regulation of cytochrome P-450-dependent microsomal drug-metabolizing enzymes by nickel, cobalt, and iron. Cobalt 89-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 334441-0 1977 Regulation of cytochrome P-450-dependent microsomal drug-metabolizing enzymes by nickel, cobalt, and iron. Iron 101-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 913025-5 1977 It is suggested that phenylbutazone and oxyphenbutazone act by inducing form of cytochrome P-450 with low activity for tolbutamide hydroxylation, whereas sulfaphenazole acts by direct inhibition of the microsomal mixed function oxidase system. Phenylbutazone 21-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 909072-0 1977 The in vitro metabolism of halothane (2-bromo-2-chloro-1,1,1-trifluoroethane) by hepatic microsomal cytochrome P-450. Halothane 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 913025-5 1977 It is suggested that phenylbutazone and oxyphenbutazone act by inducing form of cytochrome P-450 with low activity for tolbutamide hydroxylation, whereas sulfaphenazole acts by direct inhibition of the microsomal mixed function oxidase system. Oxyphenbutazone 40-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 913025-5 1977 It is suggested that phenylbutazone and oxyphenbutazone act by inducing form of cytochrome P-450 with low activity for tolbutamide hydroxylation, whereas sulfaphenazole acts by direct inhibition of the microsomal mixed function oxidase system. Tolbutamide 119-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 909072-0 1977 The in vitro metabolism of halothane (2-bromo-2-chloro-1,1,1-trifluoroethane) by hepatic microsomal cytochrome P-450. Halothane 38-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 909072-5 1977 Phenobarbital pretreatment of the animals resulted ina significant increase in both the formation of TFAA and the cytochrome P-450 content of microsomes. Phenobarbital 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 409715-1 1977 Qualitative and quantitative changes of cytochrome P-450 after chronic ethanol consumption. Ethanol 71-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 923570-0 1977 The low-spin/high-spin transition equilibrium of camphor-bound cytochrome P-450. Camphor 49-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 923570-2 1977 The spin state of camphor-bound cytochrome P-450 is shown to depend largely on medium and temperature in aqueous as well as in mixed organic buffer. Camphor 18-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 897674-1 1977 Stannous fluoride, the widely used anticaries toothpaste additive, and other tin and germanium dihalides form complexes with hemoproteins such as hepatic cytochrome P-450, hemoglobin, and peroxidase. Tin Fluorides 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-198 897674-1 1977 Stannous fluoride, the widely used anticaries toothpaste additive, and other tin and germanium dihalides form complexes with hemoproteins such as hepatic cytochrome P-450, hemoglobin, and peroxidase. Tin 77-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-198 897674-1 1977 Stannous fluoride, the widely used anticaries toothpaste additive, and other tin and germanium dihalides form complexes with hemoproteins such as hepatic cytochrome P-450, hemoglobin, and peroxidase. germanium dihalides 85-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-198 909072-6 1977 A good correlation was found between formation of TFAA and microsomal cytochrome P-450 levels. Trifluoroacetic Acid 50-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 199115-1 1977 Electron spin resonance spectroscopy as a probe of the hemin iron of cytochrome P-450: the influence of buffer composition, alcohols, and nitrogenous ligands. Iron 61-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 408351-0 1977 Heterogeneity of cytochrome P-450s induced by polychlorinated biphenyls. Polychlorinated Biphenyls 46-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 303176-7 1977 Ca2+ stimulated the reduction of cytochrome P-450 by NADPH and this was inhibited by ADP. Calcium 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 589727-0 1977 Aromatic methyl group migration and hydroxylation of p-toluidine by iron-thiol and hemin-thiol systems as a model of cytochrome P-450. 4-toluidine 53-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 589727-0 1977 Aromatic methyl group migration and hydroxylation of p-toluidine by iron-thiol and hemin-thiol systems as a model of cytochrome P-450. hemin-thiol 83-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 303176-7 1977 Ca2+ stimulated the reduction of cytochrome P-450 by NADPH and this was inhibited by ADP. NADP 53-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 303176-7 1977 Ca2+ stimulated the reduction of cytochrome P-450 by NADPH and this was inhibited by ADP. Adenosine Diphosphate 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Calcium 50-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Calcium 50-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-207 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Adenosine Diphosphate 59-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Adenosine Diphosphate 59-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-207 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Calcium 237-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Calcium 237-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-207 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Calcium 237-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Calcium 237-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-207 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Adenosine Diphosphate 307-310 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 303176-13 1977 These data have been interpreted to indicate that Ca2+ and ADP influence the relationship between adrenodoxin and cytochrome P-450 so that a complex of increased activity in the reduction of cytochrome P-450 is formed in the presence of Ca2+ and a less active complex is formed in the presence of Ca2+ plus ADP. Adenosine Diphosphate 307-310 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-207 19254-0 1977 Ionization dependence of camphor binding and spin conversion of the complex between cytochrome P-450 and camphor. Camphor 25-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 873899-1 1977 Two forms of highly purified liver microsomal cytochrome P-450, P-450LM2, and P-450LM4, have been titrated with standardized solutions of sodium dithionite under anaerobic conditions. Dithionite 138-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 19254-0 1977 Ionization dependence of camphor binding and spin conversion of the complex between cytochrome P-450 and camphor. Camphor 105-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 873899-3 1977 The present results are in disagreement with earlier dithionite titrations and reoxidation experiments which indicate that liver microsomal cytochrome P-450 is a 2-electron acceptor, but are in accord with previous potentimetric titrations and product yield data which indicate that this hemeprotein is a 1-electron acceptor. Dithionite 53-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 872430-3 1977 The cytochrome P-450 content was also measured by recording the difference absorption spectra of carbon monoxide-complexed P-450. Carbon Monoxide 97-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 198028-0 1977 [Interrelationship between the generation of oxygen anion-radicals and the reduction of artificial acceptors and cytochrome P-450 by NADPH-cytochrome c reductase]. oxygen anion-radicals 45-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 198028-4 1977 In contrast to the process of electron transfer on cytochrome c, there is a direct correlation between the rate of cytochrome P-450 reduction and the rate of adrenaline oxidation in liver microsomes. Epinephrine 158-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 198028-6 1977 On the basis of the data obtained it is suggested that the reactions of NADPH-cytochrome c reductase interaction with oxygen and artificial "anaerobic" acceptors are connected with different redox-states of flavoprotein or with different flavine coenzymes, and that the electron transport on cytochrome P-450 and directly on oxygen takes place in interrelated redox-states of flavoprotein. Oxygen 118-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 292-308 198028-6 1977 On the basis of the data obtained it is suggested that the reactions of NADPH-cytochrome c reductase interaction with oxygen and artificial "anaerobic" acceptors are connected with different redox-states of flavoprotein or with different flavine coenzymes, and that the electron transport on cytochrome P-450 and directly on oxygen takes place in interrelated redox-states of flavoprotein. 3,6-DIAMINO-10-METHYLACRIDINIUM CHLORIDE 238-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 292-308 198028-6 1977 On the basis of the data obtained it is suggested that the reactions of NADPH-cytochrome c reductase interaction with oxygen and artificial "anaerobic" acceptors are connected with different redox-states of flavoprotein or with different flavine coenzymes, and that the electron transport on cytochrome P-450 and directly on oxygen takes place in interrelated redox-states of flavoprotein. Oxygen 325-331 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 292-308 891529-0 1977 Nitrosoalkanes as Fe(II) ligands in the 455-nm-absorbing cytochrome P-450 complexes formed from nitroalkanes in reducing conditions. nitrosoalkanes 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 891529-0 1977 Nitrosoalkanes as Fe(II) ligands in the 455-nm-absorbing cytochrome P-450 complexes formed from nitroalkanes in reducing conditions. ammonium ferrous sulfate 18-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 891529-0 1977 Nitrosoalkanes as Fe(II) ligands in the 455-nm-absorbing cytochrome P-450 complexes formed from nitroalkanes in reducing conditions. nitroalkanes 96-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 193841-0 1977 Imidazole, the ligand trans to mercaptide in ferric cytochrome P-450. imidazole 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 193841-0 1977 Imidazole, the ligand trans to mercaptide in ferric cytochrome P-450. mercaptide 31-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 592792-0 1977 Ligand binding to human placental cytochrome P-450: interaction of steroids and heme-binding ligands. Steroids 67-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 888486-3 1977 The endplasmic reticulum of the hepatocytes is hypertropic but is hypoactive with respect to drug metabolism, a condition which is partly due to interference to substances retained in cholestatic livers (bile salts) with drug biotransformation and to cytochrome P-450 diminution in the microsomal membranes. Bile Acids and Salts 204-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 251-267 880239-1 1977 By using 3-amino-1,2,4-triazole, an inhibitor of haem synthesis, and 2-allyl-2-isopropylacetamide, a drug that degrades the haem moiety of cytochrome P-450, the involvement of haem in cytochrome P-450 synthesis and assembly was investigated. Amitrole 9-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 880239-1 1977 By using 3-amino-1,2,4-triazole, an inhibitor of haem synthesis, and 2-allyl-2-isopropylacetamide, a drug that degrades the haem moiety of cytochrome P-450, the involvement of haem in cytochrome P-450 synthesis and assembly was investigated. Allylisopropylacetamide 69-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 880239-1 1977 By using 3-amino-1,2,4-triazole, an inhibitor of haem synthesis, and 2-allyl-2-isopropylacetamide, a drug that degrades the haem moiety of cytochrome P-450, the involvement of haem in cytochrome P-450 synthesis and assembly was investigated. Allylisopropylacetamide 69-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-200 880239-2 1977 Phenobarbital was used to stimulate apo-(cytochrome P-450) synthesis. Phenobarbital 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 880239-7 1977 In animals receiving successive injections of phenobarbital plus 3-amino-1,2,4-triazole, compared with those receiving phenobarbital only, the holo-(cytochrome P-450) content measured spectrally shows a greater decrease than could be accounted for by the decrease in the content of the total apoprotein. Phenobarbital 46-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 880239-7 1977 In animals receiving successive injections of phenobarbital plus 3-amino-1,2,4-triazole, compared with those receiving phenobarbital only, the holo-(cytochrome P-450) content measured spectrally shows a greater decrease than could be accounted for by the decrease in the content of the total apoprotein. Amitrole 65-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 880239-7 1977 In animals receiving successive injections of phenobarbital plus 3-amino-1,2,4-triazole, compared with those receiving phenobarbital only, the holo-(cytochrome P-450) content measured spectrally shows a greater decrease than could be accounted for by the decrease in the content of the total apoprotein. Phenobarbital 119-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 880239-9 1977 This particular effect could be due to a direct interaction of 3-amino-1,2,4-triazole or its metabolites with cytochrome P-450 rather than a consequence of haem deficiency. Amitrole 63-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 880239-10 1977 Apo-(cytochrome P-450) is capable of binding to the endoplasmic reticulum in a form and at a site, which can be reconstituted with haemin to yield the functional protein. Hemin 131-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 17803-0 1977 Bioactivation of carbon tetrachloride, chloroform and bromotrichloromethane: role of cytochrome P-450. Carbon Tetrachloride 17-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 853192-0 1977 The electronic structure of chromyl chloride: a functional model for cytochrome P-450. CHROMYL CHLORIDE 28-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 884775-5 1977 Thus, in addition to its interaction with cytochrome P-450, n-octylamine causes a reduction of cytochrome b5 which subsequently becomes reoxidized. octylamine 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 17803-0 1977 Bioactivation of carbon tetrachloride, chloroform and bromotrichloromethane: role of cytochrome P-450. Bromotrichloromethane 54-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 191295-5 1977 Based on recent evidence, we conclude that it is unnecessary to postulate that ethanol is oxidized directly via cytochrome P-450. Ethanol 79-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 406056-0 1977 Separation of pure polychlorinated biphenyl isomers into two types of inducers on the basis of induction of cytochrome P-450 or P-448. Polychlorinated Biphenyls 19-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 849980-7 1977 Evidence for the presence of cytochrome P-450 in the villus fraction was obtained by measuring the carbon monoxide-dithionite difference spectrum. Carbon Monoxide 99-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 849980-7 1977 Evidence for the presence of cytochrome P-450 in the villus fraction was obtained by measuring the carbon monoxide-dithionite difference spectrum. Dithionite 115-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 881409-3 1977 On the other hand, the amount of cytochrome P-450 was depressed strongly by both cobalt treatments. Cobalt 81-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 852592-0 1977 Sub-zero temperature studies of microsomal cytochrome P-450: interaction of Fe2+ with oxygen. ammonium ferrous sulfate 76-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 881409-5 1977 The incorporations of [3H]leucine as well as 5-amino[3H]levulinate into cytochrome b5 in cobalt-treated animals were almost the same as those in the controls 5 h after injections of the radioisotopes, whereas the radioactivity of heme labelled with 5-amino[3H]levulinate in microsomal residues after trypsin digestion, which would consist mainly of cytochrome P-450, was higher in the controls than in cobalt-treated animals after 5 h. Cobalt 89-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 349-365 852592-0 1977 Sub-zero temperature studies of microsomal cytochrome P-450: interaction of Fe2+ with oxygen. Oxygen 86-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 337820-0 1977 Chronic ethanol administration induces a form of cytochrome P-450 with specific spectral and catalytic properties. Ethanol 8-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 853543-3 1977 The arylating metabolite is formed by a cytochrome P-450 dependent N-hydroxylation process. Nitrogen 67-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 19937-4 1977 Of interest are recent studies demonstrating the ability of hydrogen peroxide to initiate a cytochrome P-450 dependent peroxidatic reaction competent for supporting substrate hydroxylation. Hydrogen Peroxide 60-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 19937-5 1977 The fact that the function of cytochrome P-450 is sensitive to changes in oxygen tension establishes its role as an "oxygen sensor" for cellular metabolism. Oxygen 74-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 19937-5 1977 The fact that the function of cytochrome P-450 is sensitive to changes in oxygen tension establishes its role as an "oxygen sensor" for cellular metabolism. Oxygen 117-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 598281-0 1977 [Isolation in pure form of 20alpha, (22R)-cholesterol-hydroxylating cytochrome P-450 and some of its characteristics]. (22r)-cholesterol 36-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 140033-4 1977 Rifampicin seems to be a potent inducer of drug metabolism in humans and it causes a proliferation of the smooth endoplasmatic reticulum and an increase of cytochrome P450 content in the liver. Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-171 838934-0 1977 [Effect of catatoxic steroids on the conformation of cytochrome P-450]. Steroids 21-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 598675-0 1977 Methods for polyacrylamide gel electrophoresis of mammilian liver cytochrome P-450 in the presence of sodium dodecyl sulfact: a critique. polyacrylamide 12-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 598675-0 1977 Methods for polyacrylamide gel electrophoresis of mammilian liver cytochrome P-450 in the presence of sodium dodecyl sulfact: a critique. sodium dodecyl sulfact 102-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 1021219-1 1976 Mephenytoin, diphenylhydantoin, pheneturide, and phenobarbital produced a concentration-dependent inhibition in the binding of hexobarbital to cytochrome P-450 at the type 1 site, while sulthiame slightly potentiated, and ethosuximide did not affect the binding characteristic of hexobarbital. Mephenytoin 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 1021219-1 1976 Mephenytoin, diphenylhydantoin, pheneturide, and phenobarbital produced a concentration-dependent inhibition in the binding of hexobarbital to cytochrome P-450 at the type 1 site, while sulthiame slightly potentiated, and ethosuximide did not affect the binding characteristic of hexobarbital. Phenytoin 13-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 1021219-1 1976 Mephenytoin, diphenylhydantoin, pheneturide, and phenobarbital produced a concentration-dependent inhibition in the binding of hexobarbital to cytochrome P-450 at the type 1 site, while sulthiame slightly potentiated, and ethosuximide did not affect the binding characteristic of hexobarbital. pheneturide 32-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 1021219-1 1976 Mephenytoin, diphenylhydantoin, pheneturide, and phenobarbital produced a concentration-dependent inhibition in the binding of hexobarbital to cytochrome P-450 at the type 1 site, while sulthiame slightly potentiated, and ethosuximide did not affect the binding characteristic of hexobarbital. Phenobarbital 49-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 1021219-1 1976 Mephenytoin, diphenylhydantoin, pheneturide, and phenobarbital produced a concentration-dependent inhibition in the binding of hexobarbital to cytochrome P-450 at the type 1 site, while sulthiame slightly potentiated, and ethosuximide did not affect the binding characteristic of hexobarbital. Hexobarbital 127-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 1021219-1 1976 Mephenytoin, diphenylhydantoin, pheneturide, and phenobarbital produced a concentration-dependent inhibition in the binding of hexobarbital to cytochrome P-450 at the type 1 site, while sulthiame slightly potentiated, and ethosuximide did not affect the binding characteristic of hexobarbital. Ethosuximide 222-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 1021219-1 1976 Mephenytoin, diphenylhydantoin, pheneturide, and phenobarbital produced a concentration-dependent inhibition in the binding of hexobarbital to cytochrome P-450 at the type 1 site, while sulthiame slightly potentiated, and ethosuximide did not affect the binding characteristic of hexobarbital. Hexobarbital 280-292 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 13927-2 1976 The addition of vinyl chloride to microsomes causes a Type 1 spectra shift, similar to that seen for phenobarbital [11[ which indicates the direct involvement of a cytochrome P-450 species; this difference spectrum is characteristic of substrate binding to this type of enzyme. Vinyl Chloride 16-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-180 1001810-7 1976 Both androstenedione and 16alpha-hydroxytestosterone cause type I spectral perturbations associated with binding to cytochrome P-450 when added to placental microsomes; however, the deltaA390-420 is twice as great in response to saturating amounts of androstenedione than in response to 16alpha-hydroxytestosterone. Hydroxytestosterones 33-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 1001810-7 1976 Both androstenedione and 16alpha-hydroxytestosterone cause type I spectral perturbations associated with binding to cytochrome P-450 when added to placental microsomes; however, the deltaA390-420 is twice as great in response to saturating amounts of androstenedione than in response to 16alpha-hydroxytestosterone. Androstenedione 5-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 1001810-7 1976 Both androstenedione and 16alpha-hydroxytestosterone cause type I spectral perturbations associated with binding to cytochrome P-450 when added to placental microsomes; however, the deltaA390-420 is twice as great in response to saturating amounts of androstenedione than in response to 16alpha-hydroxytestosterone. 16-hydroxytestosterone 25-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 999697-0 1976 Platelet cytochrome P450: a possible role in arachidonate-induced aggregation. Arachidonic Acid 45-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-24 13927-2 1976 The addition of vinyl chloride to microsomes causes a Type 1 spectra shift, similar to that seen for phenobarbital [11[ which indicates the direct involvement of a cytochrome P-450 species; this difference spectrum is characteristic of substrate binding to this type of enzyme. Phenobarbital 101-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-180 184090-0 1976 Purification and characterization of adrenal cortex mitochondrial cytochrome P-450 specific for cholesterol side chain cleavage activity. Cholesterol 96-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 824157-2 1976 Cytochrome P-450 serves as the binding site for oxygen and substrate while the reductase acts as an electron carrier shuttling electrons from NADPH to cytochrome P-450. Oxygen 48-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 824157-2 1976 Cytochrome P-450 serves as the binding site for oxygen and substrate while the reductase acts as an electron carrier shuttling electrons from NADPH to cytochrome P-450. Oxygen 48-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 824157-2 1976 Cytochrome P-450 serves as the binding site for oxygen and substrate while the reductase acts as an electron carrier shuttling electrons from NADPH to cytochrome P-450. NADP 142-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 824157-2 1976 Cytochrome P-450 serves as the binding site for oxygen and substrate while the reductase acts as an electron carrier shuttling electrons from NADPH to cytochrome P-450. NADP 142-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 824157-3 1976 The phospholipid facilitates the transfer of electrons from NADPH-cytochrome c reductase to cytochrome P-450 but itself is not an electron carrier. Phospholipids 4-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 187927-0 1976 Oxidation of alpha-methyldopa and other catechols by cytochrome P-450-generated superoxide anion: possible mechanism of methyldopa hepatitis. Methyldopa 13-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 187927-0 1976 Oxidation of alpha-methyldopa and other catechols by cytochrome P-450-generated superoxide anion: possible mechanism of methyldopa hepatitis. Catechols 40-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 187927-0 1976 Oxidation of alpha-methyldopa and other catechols by cytochrome P-450-generated superoxide anion: possible mechanism of methyldopa hepatitis. Superoxides 80-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 187927-0 1976 Oxidation of alpha-methyldopa and other catechols by cytochrome P-450-generated superoxide anion: possible mechanism of methyldopa hepatitis. Methyldopa 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 12746-0 1976 Mechanism of inhibition by carbonyl cyanide m-chlorophenylhydrazone and sodium deoxycholate of cytochrome P-450-catalysed hepatic microsomal drug metabolism. Carbonyl Cyanide m-Chlorophenyl Hydrazone 27-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 12746-0 1976 Mechanism of inhibition by carbonyl cyanide m-chlorophenylhydrazone and sodium deoxycholate of cytochrome P-450-catalysed hepatic microsomal drug metabolism. Deoxycholic Acid 72-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 12746-5 1976 In the presence of 0.03-0.09% sodium deoxycholate the rate-limiting factor in p-hydroxylation of aniline is the content of cytochrome P-450. Deoxycholic Acid 30-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 184090-1 1976 Cytochrome P-450 was purified from bovine adrenal cortex mitochondria by affinity chromatography using an octylamine-substituted Sepharose column. octylamine 106-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12746-5 1976 In the presence of 0.03-0.09% sodium deoxycholate the rate-limiting factor in p-hydroxylation of aniline is the content of cytochrome P-450. aniline 97-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 184090-1 1976 Cytochrome P-450 was purified from bovine adrenal cortex mitochondria by affinity chromatography using an octylamine-substituted Sepharose column. Sepharose 129-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 184090-3 1976 The specific concentration of cytochrome P-450 in the preparation was about 5 nmol of heme per mg of protein. Heme 86-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 12746-10 1976 In the mechanism of action of carbonyl cyanide m-chlorophenylhydrazone the key step may be the electrostatic interaction of its protonated form and one of the forms of activated oxygen at the catalytic centre of cytochrome P-450. Carbonyl Cyanide m-Chlorophenyl Hydrazone 30-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 212-228 12746-10 1976 In the mechanism of action of carbonyl cyanide m-chlorophenylhydrazone the key step may be the electrostatic interaction of its protonated form and one of the forms of activated oxygen at the catalytic centre of cytochrome P-450. Oxygen 178-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 212-228 12746-11 1976 5. at least two different phospholipid-dependent hydrophobic zones are assumed to exist in the microsomal membrane, both coupled with cytochrome P-450. Phospholipids 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 12746-12 1976 One of them reveals selective sensitivity to the protonation action of carbonyl cyanide m-chlorophenylhydrazone and contains the "binding protein" for type I substrates and NADPH-cytochrome P-450 reductase; the other contains the cytochrome P-450 haem group and binding sites for type II substrates. Carbonyl Cyanide m-Chlorophenyl Hydrazone 71-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-195 184090-5 1976 Polyacrylamide gel electrophoresis of the purified cytochrome P-450 preparation treated with sodium dodecyl sulfate and mercaptoethanol showed a single major band with a molecular weight of about 60,000. polyacrylamide 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 184090-5 1976 Polyacrylamide gel electrophoresis of the purified cytochrome P-450 preparation treated with sodium dodecyl sulfate and mercaptoethanol showed a single major band with a molecular weight of about 60,000. Sodium Dodecyl Sulfate 93-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 184090-5 1976 Polyacrylamide gel electrophoresis of the purified cytochrome P-450 preparation treated with sodium dodecyl sulfate and mercaptoethanol showed a single major band with a molecular weight of about 60,000. Mercaptoethanol 120-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 184090-10 1976 After reconstitution with adrenodoxin and its reductase, this cytochrome P-450 was highly active for cholesterol desmolase with an NADPH-generating system as electron donor but was not active for steroid 11beta-hydroxylase. NADP 131-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 1068446-0 1976 Carbon monoxide binding to pentacoordinate mercaptide-heme complexes: kinetic study on models for cytochrome P-450. Carbon Monoxide 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 10855-0 1976 A highly sensitive radiometric assay for zoxazolamine hydroxylation by liver microsomal cytochrome P-450 and P-448: properties of the membrane-bound and purified reconstituted system. Zoxazolamine 41-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-114 1068446-0 1976 Carbon monoxide binding to pentacoordinate mercaptide-heme complexes: kinetic study on models for cytochrome P-450. mercaptide-heme 43-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 985554-0 1976 Polarity in aromatic hydroxylation by hemin-thiol model systems for cytochrome P-450. hemin-thiol 38-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 10902-0 1976 Stimulatory effect of FMN and methyl viologen on cytochrome P-450 dependent reduction of tertiary amine N-oxide. tertiary amine n-oxide 89-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 955110-0 1976 Ligand partitioning into membranes: its significance in determining Km and Ks values for cytochrome P-450 and other membrane bound receptors and enzymes. Potassium 75-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 954149-0 1976 A new class of inhibitory cytochrome P-450 complexes formed during metabolism: a comparison with amphetamine and SKF 525-A type complexes. Amphetamine 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 948753-0 1976 Cytochrome P-450 induction by phenobarbital and 3-methylcholanthrene in primary cultures of hepatocytes. Phenobarbital 30-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 948753-0 1976 Cytochrome P-450 induction by phenobarbital and 3-methylcholanthrene in primary cultures of hepatocytes. Methylcholanthrene 48-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 948753-1 1976 The characteristic hepatocellular changes resulting from phenobarbital administration in vivo, namely an increase in the levels of cytochrome P-450 and proliferation of membranes of the smooth endoplasmic reticulum, have been demonstrated in primary cultures of nonreplicating hepatocytes on floating collagen membranes. Phenobarbital 57-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 954149-0 1976 A new class of inhibitory cytochrome P-450 complexes formed during metabolism: a comparison with amphetamine and SKF 525-A type complexes. Proadifen 113-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 954149-1 1976 During the metabolism of p-chloroaniline, sulfanilamide, 2-methylindoline, and dapsone, complexes with cytochrome P-450 are formed which absorb maximally at 448, 450, 451, and 452 nm, respectively. 4-chloroaniline 25-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 954149-1 1976 During the metabolism of p-chloroaniline, sulfanilamide, 2-methylindoline, and dapsone, complexes with cytochrome P-450 are formed which absorb maximally at 448, 450, 451, and 452 nm, respectively. Sulfanilamide 42-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 182488-0 1976 Binding of a metyrapone spin label to microsomal cytochrome P-450. Metyrapone 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 954149-1 1976 During the metabolism of p-chloroaniline, sulfanilamide, 2-methylindoline, and dapsone, complexes with cytochrome P-450 are formed which absorb maximally at 448, 450, 451, and 452 nm, respectively. 2-Methylindoline 57-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 954149-1 1976 During the metabolism of p-chloroaniline, sulfanilamide, 2-methylindoline, and dapsone, complexes with cytochrome P-450 are formed which absorb maximally at 448, 450, 451, and 452 nm, respectively. Dapsone 79-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 954149-3 1976 Similarly, the formation is mainly a property of phenobarbital-induced cytochrome P-450 and is inhibited by comparable concentrations of metyrapone and 2-O-iodophenoxymethylimidazole. Phenobarbital 49-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 954149-3 1976 Similarly, the formation is mainly a property of phenobarbital-induced cytochrome P-450 and is inhibited by comparable concentrations of metyrapone and 2-O-iodophenoxymethylimidazole. Metyrapone 137-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 182488-1 1976 Probing of the active site of microsomal cytochrome P-450 was carried out with a spin label derived from 2-methyl-1,2-bis(3-pyridyl)-1-propanone (metyrapone). Metyrapone 105-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 182488-1 1976 Probing of the active site of microsomal cytochrome P-450 was carried out with a spin label derived from 2-methyl-1,2-bis(3-pyridyl)-1-propanone (metyrapone). Metyrapone 146-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 182488-2 1976 Its optical binding spectra to cytochrome P-450 resemble the spectra with metyrapone. Metyrapone 74-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 182488-4 1976 Since the apparent optical and EPR binding constants agree very well (Ks approximately 2-10(-5) M), and metyrapone is found to displace the spin label, we conclude, that the spin label binds to the active site of cytochrome P-450. Potassium 70-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 213-229 182488-4 1976 Since the apparent optical and EPR binding constants agree very well (Ks approximately 2-10(-5) M), and metyrapone is found to displace the spin label, we conclude, that the spin label binds to the active site of cytochrome P-450. Metyrapone 104-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 213-229 182488-6 1976 The EPR signal accounts for only 60% of the bound spin label due to its dipolar interaction with the low-spin ferric heme of cytochrome P-450. ferric heme 110-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 182488-8 1976 This supports the model that one of the pyridine nitrogens of metyrapone is coordinated to the iron of cytochrome P-450. pyridine 40-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 182488-8 1976 This supports the model that one of the pyridine nitrogens of metyrapone is coordinated to the iron of cytochrome P-450. Nitrogen 49-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 182488-8 1976 This supports the model that one of the pyridine nitrogens of metyrapone is coordinated to the iron of cytochrome P-450. Metyrapone 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 182488-8 1976 This supports the model that one of the pyridine nitrogens of metyrapone is coordinated to the iron of cytochrome P-450. Iron 95-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 939705-1 1976 The cytochrome P-450 is known to be a key enzymic component in steroid hydroxylation. Steroids 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 177476-1 1976 Models for the oxidized reaction states of cytochrome P-450 enzymes and the molecular structure of iron(III) protoporphyrin IX dimethyl ester p-nitrobenzenethiolate. iron(iii) protoporphyrin ix dimethyl ester p-nitrobenzenethiolate 99-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 956135-1 1976 The slow reduction of microsomal cytochrome P-450 by dithionite consists of an initial fast and then a slow phase. Dithionite 53-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 956135-2 1976 During reduction of aniline and azide complexes with cytochrome P-450, an intermediate spectrum developed in the fast phase and changed to that of the reduced form in the slow phase. aniline 20-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 956135-2 1976 During reduction of aniline and azide complexes with cytochrome P-450, an intermediate spectrum developed in the fast phase and changed to that of the reduced form in the slow phase. Azides 32-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 956135-6 1976 The electron spin resonance spectrum of cytochrome P450 was greatly reduced in the initial phase of reduction with dithionite. Dithionite 115-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 956135-7 1976 These results indicate that reduction of the aniline and azide complexes of cytochrome P-450 involves two steps: first reduction of cytochrome P-450 and then some changes in reduced state. aniline 45-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 956135-7 1976 These results indicate that reduction of the aniline and azide complexes of cytochrome P-450 involves two steps: first reduction of cytochrome P-450 and then some changes in reduced state. aniline 45-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 956135-7 1976 These results indicate that reduction of the aniline and azide complexes of cytochrome P-450 involves two steps: first reduction of cytochrome P-450 and then some changes in reduced state. Azides 57-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 956135-7 1976 These results indicate that reduction of the aniline and azide complexes of cytochrome P-450 involves two steps: first reduction of cytochrome P-450 and then some changes in reduced state. Azides 57-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 956135-8 1976 The aniline and cyanide difference spectra of reduced cytochrome P-450 showed peaks at 423 nm and 429 nm, respectively, while that of azide had a peak at 445 nm and a trough at 404 nm. aniline 4-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 956135-8 1976 The aniline and cyanide difference spectra of reduced cytochrome P-450 showed peaks at 423 nm and 429 nm, respectively, while that of azide had a peak at 445 nm and a trough at 404 nm. Cyanides 16-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 1275497-0 1976 Spironolactone and cytochrome P-450: impairment of steroid 21-hydroxylation in the adrenal cortex. Steroids 51-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 1257757-1 1976 Tin greatly enhances heme breakdown in kidney, thus impairing heme-dependent cellular functions, such as cytochrome P-450 mediated drug biotransformation. Tin 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 7270-0 1976 The role of cytochrome P-450 in the toxicity of fluroxene (2,2,2-trifluoroethyl vinyl ether) anaesthesia in vivo. fluroxene 48-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 1257757-1 1976 Tin greatly enhances heme breakdown in kidney, thus impairing heme-dependent cellular functions, such as cytochrome P-450 mediated drug biotransformation. Heme 62-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 7270-0 1976 The role of cytochrome P-450 in the toxicity of fluroxene (2,2,2-trifluoroethyl vinyl ether) anaesthesia in vivo. fluroxene 59-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 1278381-0 1976 A model compound for nitrosyl cytochrome P-450; further evidence for mercaptide sulfur ligation to heme. mercaptide sulfur 69-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 1262443-5 1976 These data demonstrate that metyrapone inhibits both mitochondrial reactions involved in cortisol synthesis--initial cholesterol cleavage and final 11beta-hydroxylation; these effects probably result from interference by this agent with the interaction between substrate and related cytochrome P - 450. Metyrapone 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 283-301 1278381-0 1976 A model compound for nitrosyl cytochrome P-450; further evidence for mercaptide sulfur ligation to heme. Heme 99-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 983604-0 1976 An infrared study of the carbon monoxide complexes of cytochrome P-450 and cytochrome P-420. Carbon Monoxide 25-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-91 173534-9 1975 A second effect of aniline, a type II ligand of cytochrome P-450, was to remove the g = 6 signal, suggesting that it also interacts with cytochrome P-420. aniline 19-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 998274-0 1976 On the formation of cytochrome P-450 product complexes during the metabolism of phenylalkylamines. phenylalkylamines 80-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 1008481-2 1976 In this study the ability of liver homogenates to metabolise aminopyrine and benzpyrene was correlated with hepatic cytochrome P-450 levels and with liver ultrastructure. Aminopyrine 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 1008481-2 1976 In this study the ability of liver homogenates to metabolise aminopyrine and benzpyrene was correlated with hepatic cytochrome P-450 levels and with liver ultrastructure. Benzopyrenes 77-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 1044321-0 1976 Proceedings: The role of cytochrome P-450 in carbon tetrachloride-induced hepatic injury. Carbon Tetrachloride 45-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 183224-3 1976 Both acetaminophen and furosemide decreased the concentrations of cytochrome P-450 and cytochrome b5 in microsomes, and the activity of microsomal ethylmorphine N-demethylase and aniline hydroxylase. Acetaminophen 5-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-100 183224-3 1976 Both acetaminophen and furosemide decreased the concentrations of cytochrome P-450 and cytochrome b5 in microsomes, and the activity of microsomal ethylmorphine N-demethylase and aniline hydroxylase. Furosemide 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-100 1176584-4 1975 These findings are consistent with a previously-reported spironolactone-induced destruction of the microsomal enzyme cytochrome P-450, an enzyme necessary for 17-hydroxylase and desmolase activity. Spironolactone 57-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 1209686-2 1975 A possible explanation was found when it was shown that androst-4-en-3,17-dione and testosterone could displace bound carbon monoxide from human placental microsomal cytochrome P-450. Androstenedione 56-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-182 1209686-2 1975 A possible explanation was found when it was shown that androst-4-en-3,17-dione and testosterone could displace bound carbon monoxide from human placental microsomal cytochrome P-450. Testosterone 84-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-182 1209686-2 1975 A possible explanation was found when it was shown that androst-4-en-3,17-dione and testosterone could displace bound carbon monoxide from human placental microsomal cytochrome P-450. Carbon Monoxide 118-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-182 6092-4 1975 VMAX for the hydroxylation of DMA calculated as per 1 nmol of the cytochrome P--450 was higher in the cells than in the microsomes. N,N-dimethylaniline 30-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-83 1216968-0 1975 Proceedings: The effects of adducts and the nature of activated oxygen for cytochrome P-450 catalyzed hydroxylation reactions. Oxygen 64-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 1052431-0 1975 Effect of cobalt on microsomal cytochrome P-450: differences between liver and intestinal mucosa. Cobalt 10-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 1160997-0 1975 Early role during chemical evolution for cytochrome P450 in oxygen detoxification. Oxygen 60-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 1201750-3 1975 On the basis of binding studies with ethyl isocyanide, degradation of cytochrome P-450 to P-420, redox potential, aniline binding, and relative rates of reduction by NADPH and NADH, it is suggested that the cytochrome P-450 system is analogous to that mammalian microsomes. ethyl isocyanide 37-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 166885-0 1975 Nitric oxide complexes of cytochrome P-450. Nitric Oxide 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 240656-0 1975 Interaction of aromatic nitro compounds with reduced hepatic microsomal cytochrome P-450. aromatic nitro compounds 15-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 240656-5 1975 There is apparent mutual inhibition of binding of carbon monoxide, metyrapone, and nitro compounds with reduced cytochrome P-450, since addition of metyrapone or carbon monoxide to reference and sample cuvettes does not alter qualitative aspects of the nitro binding spectra, but only its magnitude. Carbon Monoxide 50-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 240656-5 1975 There is apparent mutual inhibition of binding of carbon monoxide, metyrapone, and nitro compounds with reduced cytochrome P-450, since addition of metyrapone or carbon monoxide to reference and sample cuvettes does not alter qualitative aspects of the nitro binding spectra, but only its magnitude. Metyrapone 67-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 1201750-3 1975 On the basis of binding studies with ethyl isocyanide, degradation of cytochrome P-450 to P-420, redox potential, aniline binding, and relative rates of reduction by NADPH and NADH, it is suggested that the cytochrome P-450 system is analogous to that mammalian microsomes. aniline 114-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 240656-5 1975 There is apparent mutual inhibition of binding of carbon monoxide, metyrapone, and nitro compounds with reduced cytochrome P-450, since addition of metyrapone or carbon monoxide to reference and sample cuvettes does not alter qualitative aspects of the nitro binding spectra, but only its magnitude. nitro 83-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 240656-6 1975 From the relative KS values, it is concluded that metyrapone and carbon monoxide interact with reduced cytochrome P-450 more tenaciously than nitro compounds. Potassium 18-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 1201750-3 1975 On the basis of binding studies with ethyl isocyanide, degradation of cytochrome P-450 to P-420, redox potential, aniline binding, and relative rates of reduction by NADPH and NADH, it is suggested that the cytochrome P-450 system is analogous to that mammalian microsomes. NADP 166-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 240656-6 1975 From the relative KS values, it is concluded that metyrapone and carbon monoxide interact with reduced cytochrome P-450 more tenaciously than nitro compounds. Metyrapone 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 1201750-3 1975 On the basis of binding studies with ethyl isocyanide, degradation of cytochrome P-450 to P-420, redox potential, aniline binding, and relative rates of reduction by NADPH and NADH, it is suggested that the cytochrome P-450 system is analogous to that mammalian microsomes. NADP 166-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 240656-6 1975 From the relative KS values, it is concluded that metyrapone and carbon monoxide interact with reduced cytochrome P-450 more tenaciously than nitro compounds. Carbon Monoxide 65-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 1201750-3 1975 On the basis of binding studies with ethyl isocyanide, degradation of cytochrome P-450 to P-420, redox potential, aniline binding, and relative rates of reduction by NADPH and NADH, it is suggested that the cytochrome P-450 system is analogous to that mammalian microsomes. NAD 176-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-223 1233229-5 1975 The acceleration of oestrogen hydroxylation by rifampicin was paralleled by an increase in microsomal cytochrome P-450, and also by microsomal reduction of rifampicin-quinone, a reactive metabolite of rifampicin. Rifampin 47-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 1902-2 1975 In oxidation of NADP.H2 there were at least three point of molecular O2 reduction: NADP.H2-specific flavoprotein, Fe2+ participating in reactions of peroxidation of unsaturated fatty acids and cytochrome P-450. NADP 16-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-209 1142995-0 1975 Interactions of steroids with human placental cytochrome P-450 in the presence of carbon monoxide. Steroids 16-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 1142995-0 1975 Interactions of steroids with human placental cytochrome P-450 in the presence of carbon monoxide. Carbon Monoxide 82-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 806264-0 1975 Metobolism of chlorobenzene with hepatic microsomes and solubilized cytochrome P-450 systems. chlorobenzene 14-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 1902-2 1975 In oxidation of NADP.H2 there were at least three point of molecular O2 reduction: NADP.H2-specific flavoprotein, Fe2+ participating in reactions of peroxidation of unsaturated fatty acids and cytochrome P-450. Hydrogen 21-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-209 1902-2 1975 In oxidation of NADP.H2 there were at least three point of molecular O2 reduction: NADP.H2-specific flavoprotein, Fe2+ participating in reactions of peroxidation of unsaturated fatty acids and cytochrome P-450. Oxygen 69-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-209 1902-2 1975 In oxidation of NADP.H2 there were at least three point of molecular O2 reduction: NADP.H2-specific flavoprotein, Fe2+ participating in reactions of peroxidation of unsaturated fatty acids and cytochrome P-450. NADP 83-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-209 1902-3 1975 Efficiency of cytochrome P-450 inhibitors could not be evaluated by polarography as in the pathway several sites of molecular O2 activation were observed. Oxygen 126-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 1131251-0 1975 Purification of cytochrome P-450 from bovin adrenocortical mitochondria by an "aniline-sepharose" and the properties. aniline 79-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 1131251-0 1975 Purification of cytochrome P-450 from bovin adrenocortical mitochondria by an "aniline-sepharose" and the properties. Sepharose 87-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 1186237-0 1975 The role of cholesterol and cytochrome P-450 in the cholesterol side chain cleavage reaction in adrenal cortex and corpora lutea. Cholesterol 52-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 1133375-1 1975 Possible relationship to cytochrome P-450 enzymes and the structure of (p-nitrobenzenethiolato)iron(III) protoporphyrin IX dimethyl ester. (p-nitrobenzenethiolato)iron(iii) protoporphyrin ix dimethyl ester 71-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 234439-5 1975 Cytochrome P-450 is recovered in a floating pellet after the addition of 25% ammonium sulfate followed by centrifugation, whereas cytochrome P-450I remains in the 25% ammonium sulfate supernatant fluid. Ammonium Sulfate 77-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 234439-6 1975 Cytochrome P-450 is purified further by Sephadez G-200 and DEAE-Sephadex A-50 column chromatography, which also allows the isolation of cytochrome b5 and NADPH-dependent cytochrome P-450 reductase in good yields and with little cross-contamination. sephadez g-200 40-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 234439-6 1975 Cytochrome P-450 is purified further by Sephadez G-200 and DEAE-Sephadex A-50 column chromatography, which also allows the isolation of cytochrome b5 and NADPH-dependent cytochrome P-450 reductase in good yields and with little cross-contamination. 2-diethylaminoethanol 59-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 234439-16 1975 The cytochrome P-450 aggregate is dissociated by 6 M urea, but cytochrome P-450I remains as such. Urea 53-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 4374132-0 1974 Reduced nicotinamide adenine dinucleotide-cytochrome b5 reductase and cytochrome b5 as electron carriers in NADH-supported cytochrome P-450 -dependent enzyme activities in liver microsomes. NAD 108-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 805803-0 1975 Involvement of cytochrome P-450 in the intracellular formation of lipid peroxides. Lipid Peroxides 66-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 1120111-4 1975 (1) The hydrophilic sulfate group directs the steroid molecule so that it only interacts with the active site of cytochrome P-450 with its non-sulfurylated, hydrophobic end. Sulfates 20-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 1120111-4 1975 (1) The hydrophilic sulfate group directs the steroid molecule so that it only interacts with the active site of cytochrome P-450 with its non-sulfurylated, hydrophobic end. Steroids 46-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 1120111-5 1975 (2) The sulfate group interacts with the enzyme surface resulting in exposure of a slightly different part of the hydrophobic end of the substrate to the active site of cytochrome P-450 than when the same end of the free steroid is exposed to the active site of the enzyme. Sulfates 8-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 1120111-5 1975 (2) The sulfate group interacts with the enzyme surface resulting in exposure of a slightly different part of the hydrophobic end of the substrate to the active site of cytochrome P-450 than when the same end of the free steroid is exposed to the active site of the enzyme. Steroids 221-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 1139001-1 1975 The absence of correlation between the effect of aniline and aminoantipyrine derivatives on cytochrome P-450 reduction rate and its oxidation rate draw to the conclusion that the reductase reaction is not a limiting step of hydroxylation for all substrates. Ampyrone 61-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 1116950-5 1975 Subtraction of the nitrogen values from the carbon monoxide values, after allowing for an absorption shift, gives the absolute spectrum of cytochrome P450. Nitrogen 19-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 1116950-5 1975 Subtraction of the nitrogen values from the carbon monoxide values, after allowing for an absorption shift, gives the absolute spectrum of cytochrome P450. Carbon Monoxide 44-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 1116950-8 1975 The content of cytochrome P450 in animals fed with phenobarbitone was 2.4 times greater than in control animals. Phenobarbital 51-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-30 4550000-0 1974 Spironolactone and cytochrome P-450: impairment of steroid hydroxylation in the adrenal cortex. Steroids 51-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 4371680-0 1974 Spectral and catalytic properties of cytochrome P-450 from a diazinon-resistant housefly strain. Diazinon 61-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 4531003-5 1974 We have also shown that the molar magnetic ellipticity for reduced + CO treated cytochrome P-450 of Pseudomonas grown on camphor is of similar value to that of reduced + CO treated microsomal P-450 and P-448. Camphor 121-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 4432244-0 1974 Increased antitumor activity of cyclophosphamide (Endoxan) following pretreatment with inducer of drug-metabolizing enzymes (cytochrome P-450). Cyclophosphamide 32-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 4432244-0 1974 Increased antitumor activity of cyclophosphamide (Endoxan) following pretreatment with inducer of drug-metabolizing enzymes (cytochrome P-450). Cyclophosphamide 50-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 4153511-0 1974 The formation of complexes absorbing at 455 nm from cytochrome P-450 and metabloites of compounds related to SKF 525-A. Proadifen 109-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 4831127-0 1974 Spironolactone and testicular cytochrome P-450: decreased testosterone formation in several species and changes in hepatic drug metabolism. Testosterone 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 4153124-0 1974 The influence of cytochrome P-450 induction on the metabolic formation of 455-NM complexes from amphetamines. Amphetamines 96-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 4364009-0 1974 Phospholipids of adrenal cortex mitochondria and the steroid hydroxylases: the lipid-environment of cytochrome P-450. Phospholipids 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 4151581-0 1974 Redox properties of the reduced nicotinamide adenine dinucleotide phosphate-cytochrome P-450 and reduced nicotinamide adenine dinucleotide-cytochrome b5 reductases. NAD 32-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 4136627-0 1974 Detection of cytochrome P-450 in subcellular fractions of Endomycopsislipolytica grown on n-hexadecane. n-hexadecane 90-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 4204348-0 1974 Differences in inducibility of cutaneous and hepatic drug metabolizing enzymes and cytochrome P-450 by polychlorinated biphenyls and 1,1,1-trichloro-2,2-bis(p-chlorophenyl)ethane (DDT). Polychlorinated Biphenyls 103-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 4522784-1 1974 Induction of hepatic microsomal cytochrome P-450 and ethylmorphine N-demethylase activity by phenobarbital requires de novo synthesis of mRNA. Phenobarbital 93-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-80 4828525-0 1974 The effect of phenobarbital, 3-methylcholanthrene, 3,4-benzpyrene, and pregnenolone-16 alpha-carbonitrile on microsomal heme oxygenase and splenic cytochrome P-450. Pregnenolone Carbonitrile 71-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 4204348-0 1974 Differences in inducibility of cutaneous and hepatic drug metabolizing enzymes and cytochrome P-450 by polychlorinated biphenyls and 1,1,1-trichloro-2,2-bis(p-chlorophenyl)ethane (DDT). DDT 133-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 4204348-0 1974 Differences in inducibility of cutaneous and hepatic drug metabolizing enzymes and cytochrome P-450 by polychlorinated biphenyls and 1,1,1-trichloro-2,2-bis(p-chlorophenyl)ethane (DDT). DDT 180-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 4721048-1 1973 Rebinding of carbon monoxide to myoglobin and to cytochrome P-450 after removal by a light flash occurs down to 50 degrees K for myoglobin and 25 degrees K for cytochrome P-450 in glycerol-water solution. Carbon Monoxide 13-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 4149283-0 1973 The role of cytochrome P-450 in cholesterol biosynthesis. Cholesterol 32-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 4721048-1 1973 Rebinding of carbon monoxide to myoglobin and to cytochrome P-450 after removal by a light flash occurs down to 50 degrees K for myoglobin and 25 degrees K for cytochrome P-450 in glycerol-water solution. Carbon Monoxide 13-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 4721048-1 1973 Rebinding of carbon monoxide to myoglobin and to cytochrome P-450 after removal by a light flash occurs down to 50 degrees K for myoglobin and 25 degrees K for cytochrome P-450 in glycerol-water solution. Glycerol 180-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 4721048-1 1973 Rebinding of carbon monoxide to myoglobin and to cytochrome P-450 after removal by a light flash occurs down to 50 degrees K for myoglobin and 25 degrees K for cytochrome P-450 in glycerol-water solution. Glycerol 180-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 4721048-1 1973 Rebinding of carbon monoxide to myoglobin and to cytochrome P-450 after removal by a light flash occurs down to 50 degrees K for myoglobin and 25 degrees K for cytochrome P-450 in glycerol-water solution. Water 189-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 4721048-1 1973 Rebinding of carbon monoxide to myoglobin and to cytochrome P-450 after removal by a light flash occurs down to 50 degrees K for myoglobin and 25 degrees K for cytochrome P-450 in glycerol-water solution. Water 189-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 4733995-0 1973 Role of cytochrome P-450 in carbon tetrachloride activation and CCl 4-induced necrosis. Carbon Tetrachloride 28-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 4741599-0 1973 Some spectral properties of cytochrome P-450 from microsomes isolated from control, phenobarbital- and naphthalene-treated houseflies. Phenobarbital 84-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 4741599-0 1973 Some spectral properties of cytochrome P-450 from microsomes isolated from control, phenobarbital- and naphthalene-treated houseflies. naphthalene 103-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 4122821-0 1973 The role of cytochrome P-450 in N-hydroxylation of 2-acetylaminofluorene. Nitrogen 32-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 4122821-0 1973 The role of cytochrome P-450 in N-hydroxylation of 2-acetylaminofluorene. 2-Acetylaminofluorene 51-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 4146295-0 1973 Iron-dependent loss of liver cytochrome P-450 haem in vivo and in vitro. Iron 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 4149394-0 1973 Breakdown of cytochrome P-450 heme by secobarbital and other allyl-containing barbiturates. Heme 30-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 4155922-0 1973 The role of cytochrome P-450 and P-448 in drug and steroid hydroxylations. Steroids 51-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-38 4375425-0 1973 Testicular cytochrome P-450 and iron-sulfur protein as related to steroid metabolism. Steroids 66-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 4149394-0 1973 Breakdown of cytochrome P-450 heme by secobarbital and other allyl-containing barbiturates. Barbiturates 78-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 4149397-0 1973 The role of cytochrome P-450 in the N-oxidation of individual amines. Amines 62-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 4650623-0 1972 A reconstituted microsomal enzyme system that converts naphthalene to trans-1,2-dihydroxy-1,2-dihydronaphthalene via naphthalene-1,2-oxide: presence of epoxide hydrase in cytochrome P-450 and P-448 fractions. naphthalene 55-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-197 4267597-0 1973 Active centers of microsomal cytochrome P-450 determined with metyrapone by mutural depletion system kinetics. Metyrapone 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 4650623-0 1972 A reconstituted microsomal enzyme system that converts naphthalene to trans-1,2-dihydroxy-1,2-dihydronaphthalene via naphthalene-1,2-oxide: presence of epoxide hydrase in cytochrome P-450 and P-448 fractions. 1,2-dihydroxy-1,2-dihydronaphthalene 70-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-197 4650623-0 1972 A reconstituted microsomal enzyme system that converts naphthalene to trans-1,2-dihydroxy-1,2-dihydronaphthalene via naphthalene-1,2-oxide: presence of epoxide hydrase in cytochrome P-450 and P-448 fractions. naphthalene 1,2-oxide 117-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-197 11946722-0 1972 On the participation of cytochrome P-450 in the formation of 16, 17-dihydroxylated C(19) steroids from 16-dehydro-C(19) steroids. 16, 17-dihydroxylated c 61-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 11946722-0 1972 On the participation of cytochrome P-450 in the formation of 16, 17-dihydroxylated C(19) steroids from 16-dehydro-C(19) steroids. Steroids 89-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 11946722-0 1972 On the participation of cytochrome P-450 in the formation of 16, 17-dihydroxylated C(19) steroids from 16-dehydro-C(19) steroids. 16-dehydro-c 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 11946722-0 1972 On the participation of cytochrome P-450 in the formation of 16, 17-dihydroxylated C(19) steroids from 16-dehydro-C(19) steroids. Steroids 120-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 4401153-2 1972 Role of the cytochrome P-450 and P-448 fractions in drug and steroid hydroxylations. Steroids 61-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-38 5051307-0 1972 Decrease in hepatic cytochrome P-450 and catalase following allylisopropyl acetamide: the effect of concomitant hemin administration. Allylisopropylacetamide 60-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-49 5076665-0 1972 Is the liver cytochrome P-450-linked biotransformation system lecithin-dependent? Lecithins 62-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 5013819-0 1972 Spectral studies of the binding of O,O-diethyl p-nitrophenylphosphorothionate (parathion) to cytochrome P-450. o,o-diethyl p-nitrophenylphosphorothionate 35-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 5008942-0 1972 Decreased levels of cytochrome P-450 and catalase in hepatic porphyria caused by substituted acetamides and barbiturates. Acetamides 93-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-49 5013819-0 1972 Spectral studies of the binding of O,O-diethyl p-nitrophenylphosphorothionate (parathion) to cytochrome P-450. Parathion 79-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 5008942-0 1972 Decreased levels of cytochrome P-450 and catalase in hepatic porphyria caused by substituted acetamides and barbiturates. Barbiturates 108-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-49 4399944-0 1971 Conversion of agroclavine by mammalian cytochrome P-450. agroclavine 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 4335684-0 1971 Cytochrome P-450 from the adrenal cortex: interaction of steroids and the hydroxylation reaction. Steroids 57-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 5144763-0 1971 The binding of metyrapone to cytochrome P-450 and its inhibitory action on liver microsomal mixed-function oxidase reactions. Metyrapone 15-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 5144765-0 1971 Model systems for mono-oxygenases, with results with iron(II)-sulphur complexes as models for cytochrome P-450. ammonium ferrous sulfate 53-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 5116661-0 1971 Formation rates of the two ethyl isocyanide compounds of liver cytochrome P-450 at low temperatures. ethyl isocyanide 27-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 5098347-0 1971 [Binding of carbon tetrachloride to reduced microsomal cytochrome P-450 and to heme]. Carbon Tetrachloride 12-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 4400522-0 1972 Incorporation of radioactive- -aminolevulinic acid into microsomal cytochrome P 450 : selective breakdown of the hemoprotein by allylisopropylacetamide and carbon tetrachloride. radioactive- -aminolevulinic acid 17-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 4400522-0 1972 Incorporation of radioactive- -aminolevulinic acid into microsomal cytochrome P 450 : selective breakdown of the hemoprotein by allylisopropylacetamide and carbon tetrachloride. Allylisopropylacetamide 128-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 4400522-0 1972 Incorporation of radioactive- -aminolevulinic acid into microsomal cytochrome P 450 : selective breakdown of the hemoprotein by allylisopropylacetamide and carbon tetrachloride. Carbon Tetrachloride 156-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 4404917-0 1972 [Formation of chloroform from carbon tetrachloride in liver microsomes, lipid peroxidation and destruction of cytochrome P-450]. Chloroform 14-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-127 4570969-0 1972 The binding of metyrapone to cytochrome P-450 and its inhibitory action on microsomal hepatic mixed function oxidation reactions. Metyrapone 15-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 5118111-0 1971 Species differences in the effect of morphine administration or adrenalectomy on the substrate interactions with cytochrome P-450 and drug oxidations by liver microsomes. Morphine 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 4397379-0 1971 Effect of temperature on the relationship between hepatic microsomal cytochrome P-450 reduction and ethylmorphine demethylation. Ethylmorphine 100-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 4099847-0 1971 Properties of cytochrome P-450 as affected by environmental factors: qualitative changes due to administration of polycyclic hydrocarbons. Hydrocarbons, Cyclic 114-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 5289597-0 1970 Some chemical properties of cytochrome P-450 and its carbon monoxide compound (P-450.CO). Carbon Monoxide 53-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 5497445-0 1970 Incorporation of radioactive iron into cytochrome b5 and cytochrome P-450 of liver microsomes. Iron 29-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 5535939-0 1970 Effect of adrenalectomy on the substrate interaction with cytochrome P-450 in the hydroxylation of steroid hormones by liver microsomes. Steroids 99-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 11947411-0 1970 Rapid loss of cytochrome P-450 and haem caused in the liver microsomes by the porphyrogenic agent 2-allyl-2-isopropylacetamide. Allylisopropylacetamide 98-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 4391249-0 1969 Cytochrome P-450 in steroid biogenesis. Steroids 20-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 5360680-0 1969 Cytochrome P-450 in the omega-oxidation of fatty acids. Fatty Acids 43-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 4390185-0 1969 The role of heme synthesis during the induction of hepatic microsomal cytochrome P-450 and drug metabolism produced by benzpyrene. Heme 12-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 5433592-0 1970 In vivo interaction of metyrapone with adrenal cortical mitochondrial cytochrome P-450. Metyrapone 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 4390187-0 1969 Hydroxylation of benzphetamine and other drugs by a solubilized form of cytochrome P-450 from liver microsomes: lipid requirement for drug demethylation. Benzphetamine 17-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 4185033-0 1969 The role of cytochrome P-450 in the mechanism of inhibition of steroid 11 beta-hydroxylation by dicumarol. Steroids 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 5796288-0 1969 A model for the interaction of cytochrome P-450 with carbon monoxide. Carbon Monoxide 53-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 4185033-0 1969 The role of cytochrome P-450 in the mechanism of inhibition of steroid 11 beta-hydroxylation by dicumarol. Dicumarol 96-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 5805284-0 1969 The interaction of metopirone with adrenal mitochondrial cytochrome P-450. Metyrapone 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 5725573-0 1968 Mechanism of p-nitrobenzoate reduction in liver: the possible role oc cytochrome P-450 in liver microsomes. p-nitrobenzoate 13-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 4385526-0 1968 The electron spin resonance and absorption spectra of microsomal cytochrome P-450 and its isocyanide complexes. Cyanides 90-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 33932713-0 2021 Probing cytochrome P450 (CYP) bioactivation with chloromethylindoline bioprecursors derived from the duocarmycin family of compounds. chloromethylindoline 49-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 5688265-0 1968 Effect of partial hepatectomy of the responsiveness of microsomal enzymes and cytochrome P-450 to phenobarbital or 3-methylcholanthrene. Phenobarbital 98-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 5688265-0 1968 Effect of partial hepatectomy of the responsiveness of microsomal enzymes and cytochrome P-450 to phenobarbital or 3-methylcholanthrene. Methylcholanthrene 115-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 33932713-3 2021 In this study, we have explored seco-OH-chloromethylindoline (CI) duocarmycin-based bioprecursors for their potential for cytochrome P450 (CYP)-mediated cancer cell kill. morin 62-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 33932713-3 2021 In this study, we have explored seco-OH-chloromethylindoline (CI) duocarmycin-based bioprecursors for their potential for cytochrome P450 (CYP)-mediated cancer cell kill. morin 62-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 33932713-0 2021 Probing cytochrome P450 (CYP) bioactivation with chloromethylindoline bioprecursors derived from the duocarmycin family of compounds. chloromethylindoline 49-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 33932713-3 2021 In this study, we have explored seco-OH-chloromethylindoline (CI) duocarmycin-based bioprecursors for their potential for cytochrome P450 (CYP)-mediated cancer cell kill. seco-oh-chloromethylindoline 32-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 33932713-3 2021 In this study, we have explored seco-OH-chloromethylindoline (CI) duocarmycin-based bioprecursors for their potential for cytochrome P450 (CYP)-mediated cancer cell kill. seco-oh-chloromethylindoline 32-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 28520368-5 2012 Clozapine is metabolized in the liver by the cytochrome P450 (CYP) system of enzymes. Clozapine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 33652071-0 2021 Identification of human cytochrome P450 isozymes involved in the oxidative metabolism of carfentanil. carfentanil 89-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 33652071-2 2021 In the present study, the human cytochrome P450 (CYP) isozymes catalyzing the oxidative metabolism of carfentanil were investigated. carfentanil 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 33652071-2 2021 In the present study, the human cytochrome P450 (CYP) isozymes catalyzing the oxidative metabolism of carfentanil were investigated. carfentanil 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 28520368-6 2012 CYP1A2 is the main CYP isoform in clozapine metabolism and CYP1A2 activity is an important determinant of clozapine dose (4). Clozapine 34-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 28520368-6 2012 CYP1A2 is the main CYP isoform in clozapine metabolism and CYP1A2 activity is an important determinant of clozapine dose (4). Clozapine 106-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 28520368-5 2012 Clozapine is metabolized in the liver by the cytochrome P450 (CYP) system of enzymes. Clozapine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 28520368-7 2012 Other CYP enzymes involved in clozapine metabolism include CYP2D6 and CYP3A4. Clozapine 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 34048999-1 2021 Gilteritinib is primarily metabolized via cytochrome P450 (CYP). gilteritinib 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 34048999-1 2021 Gilteritinib is primarily metabolized via cytochrome P450 (CYP). gilteritinib 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 33982687-1 2021 Cytochrome-P450-reductase transfers electrons to cytochrome-P450 through its flavin mononucleotide binding domain (FBD). Flavin Mononucleotide 77-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 33610566-2 2021 However, its glucuronidation, glucuronides excretion, and drug-drug interaction (DDI) involving in human cytochrome P450 (CYP), UDP-glucuronosyltransferase (UGT) enzymes, and efflux transporters (BCRP and MRPs) remains unclear so far. Glucuronides 30-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 33934248-0 2021 Transcriptomic analysis of saffron at different flowering stages using RNA sequencing uncovers cytochrome P450 genes involved in crocin biosynthesis. crocin 129-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 33934248-2 2021 This study aimed to screen differentially expressed genes (DEGs) in saffron at different flowering stages and identify cytochrome P450 (CYP) genes involved in crocin biosynthesis. crocin 159-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-134 33934248-2 2021 This study aimed to screen differentially expressed genes (DEGs) in saffron at different flowering stages and identify cytochrome P450 (CYP) genes involved in crocin biosynthesis. crocin 159-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-139 6541651-2 1984 In spite of numerous spectroscopic similarities between chloroperoxidase and cytochrome P-450 (P-450) which suggest endogenous cysteinate axial ligation in chloroperoxidase as has been established for P-450, assignment of the endogenous axial ligand of chloroperoxidase has remained controversial since no available free sulfhydryl groups have been detected in chemical studies of chloroperoxidase. cysteinate 127-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 33610566-0 2021 Potential Determinants for Metabolic Fates and Inhibitory Effects of Isobavachalcone Involving in Human Cytochrome P450, UDP-Glucuronosyltransferase Enzymes, and Efflux Transporters. isobavachalcone 69-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 33610566-2 2021 However, its glucuronidation, glucuronides excretion, and drug-drug interaction (DDI) involving in human cytochrome P450 (CYP), UDP-glucuronosyltransferase (UGT) enzymes, and efflux transporters (BCRP and MRPs) remains unclear so far. Glucuronides 30-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 33748959-2 2021 Their narrow therapeutic index and metabolism through cytochrome p450 (CYP) enzymes can result in a drug interaction when used concomitantly with rifamycins. rifamycin S 146-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 33748959-2 2021 Their narrow therapeutic index and metabolism through cytochrome p450 (CYP) enzymes can result in a drug interaction when used concomitantly with rifamycins. rifamycin S 146-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 33748959-4 2021 OBJECTIVE: To examine literature which evaluates the concomitant use of opioids and rifamycins with clinically-relevant CYP-inducing properties METHODS: A systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria was performed. rifamycin S 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 33987428-12 2021 Circumstantial evidence suggests that reactive oxygen species (ROS) generated by cytochrome P450 could be responsible for the initial steps of injury, but details are unknown. Reactive Oxygen Species 38-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 33890704-1 2021 We investigated the effect of deglucuronidation on the plasma concentration of the constituents of the Basel phenotyping cocktail and on the interpretation of the phenotyping results under basal conditions and after cytochrome P450 (CYP) induction with metamizole. Dipyrone 253-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-231 33719395-2 2021 Several of these PAs are genotoxic and carcinogenic, primarily in the liver, upon cytochrome P450 (CYP)-catalyzed activation into reactive (pyrrolic and pyrrole-like) metabolites. Pyrroles 153-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 33719395-2 2021 Several of these PAs are genotoxic and carcinogenic, primarily in the liver, upon cytochrome P450 (CYP)-catalyzed activation into reactive (pyrrolic and pyrrole-like) metabolites. Pyrroles 153-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-102 33728909-2 2021 We aimed to investigate the inhibitory effect of binimetinib on cytochrome P450 using human liver microsomes. binimetinib 49-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 33592181-1 2021 Human cytochrome P450 enzymes (CYPs or P450s) are known to be reduced by their electron transfer partners in the absence of substrate and in turn to reduce other acceptor molecules such as molecular oxygen, thereby creating superoxide anions (O2- ). Oxygen 199-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 33592181-1 2021 Human cytochrome P450 enzymes (CYPs or P450s) are known to be reduced by their electron transfer partners in the absence of substrate and in turn to reduce other acceptor molecules such as molecular oxygen, thereby creating superoxide anions (O2- ). Superoxides 224-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 33592181-1 2021 Human cytochrome P450 enzymes (CYPs or P450s) are known to be reduced by their electron transfer partners in the absence of substrate and in turn to reduce other acceptor molecules such as molecular oxygen, thereby creating superoxide anions (O2- ). Oxygen 243-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 33935788-14 2021 The correlation analysis between CYP protein abundance and CYP enzyme activity showed that CYP3A22 protein abundance correlated clearly with the metabolism of midazolam at PND 7. Midazolam 159-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 33935788-14 2021 The correlation analysis between CYP protein abundance and CYP enzyme activity showed that CYP3A22 protein abundance correlated clearly with the metabolism of midazolam at PND 7. Midazolam 159-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 33936006-1 2021 Cytochrome P450 enzymes, or P450s, are haem monooxygenases renowned for their ability to insert one atom from molecular oxygen into an exceptionally broad range of substrates while reducing the other atom to water. Oxygen 48-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 33936006-1 2021 Cytochrome P450 enzymes, or P450s, are haem monooxygenases renowned for their ability to insert one atom from molecular oxygen into an exceptionally broad range of substrates while reducing the other atom to water. Water 208-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 33987428-12 2021 Circumstantial evidence suggests that reactive oxygen species (ROS) generated by cytochrome P450 could be responsible for the initial steps of injury, but details are unknown. Reactive Oxygen Species 63-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 33538960-9 2021 This complicates the drug-disease interaction profile of the patients as both the investigational drugs (e.g., remdesivir, dexamethasone) and the agents for comorbidities can be affected by compromised CYP-mediated hepatic metabolism. Dexamethasone 123-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-205 33400885-1 2021 INTRODUCTION: Cytochrome P450 (CYP) metabolizes vital endogenous (steroids, vitamins) and exogenous (drugs, toxins) substrates. Steroids 66-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 33400885-1 2021 INTRODUCTION: Cytochrome P450 (CYP) metabolizes vital endogenous (steroids, vitamins) and exogenous (drugs, toxins) substrates. Steroids 66-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 33400885-6 2021 An opposite role of the hypothalamic paraventricular and arcuate nuclei and 5-HT receptors present therein in the regulation of CYP is described. Serotonin 76-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 33189850-1 2021 Cytochrome P450 (P450) 11B1 and 11B2 both catalyze the 11beta-hydroxylation of 11-deoxycorticosterone and the subsequent 18-hydroxylation of the product. Desoxycorticosterone 79-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-27 33570057-5 2021 Rhamnetin treatment decreased expression of the drug resistance-related downstream genes of PXR (cyp3a4 [cytochrome P-450] or mdr-1 [multi-drug resistance 1]), which mediate the metabolism or elimination of sorafenib in HCC cells. rhamnetin 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 33570057-5 2021 Rhamnetin treatment decreased expression of the drug resistance-related downstream genes of PXR (cyp3a4 [cytochrome P-450] or mdr-1 [multi-drug resistance 1]), which mediate the metabolism or elimination of sorafenib in HCC cells. Sorafenib 207-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 33545101-2 2021 We demonstrate the nascent approach of site-specific 2D IR spectroscopy to investigate the archetypical cytochrome P450, P450cam, to better delineate the mechanism of the lower regioselectivity of hydroxylation of the substrate norcamphor in comparison to the native substrate camphor. norcamphor 228-238 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 33545101-2 2021 We demonstrate the nascent approach of site-specific 2D IR spectroscopy to investigate the archetypical cytochrome P450, P450cam, to better delineate the mechanism of the lower regioselectivity of hydroxylation of the substrate norcamphor in comparison to the native substrate camphor. Camphor 231-238 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 33212179-6 2021 The highest activity was exhibited by methyl ophiopogonanone B and ginsenoside F2, indicating that they are CYP inducers. methylophiopogonanone B 38-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 33212179-6 2021 The highest activity was exhibited by methyl ophiopogonanone B and ginsenoside F2, indicating that they are CYP inducers. ginsenoside F2 67-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 33538960-9 2021 This complicates the drug-disease interaction profile of the patients as both the investigational drugs (e.g., remdesivir, dexamethasone) and the agents for comorbidities can be affected by compromised CYP-mediated hepatic metabolism. remdesivir 111-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-205 33125290-0 2021 Human Mass Balance, Metabolism, and Cytochrome P450 Phenotyping of Lusutrombopag. lusutrombopag 67-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 32918831-0 2021 Evaluation of Potential Drug-Drug Interaction Risk of Pexidartinib With Substrates of Cytochrome P450 and P-Glycoprotein. pexidartinib 54-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 33747646-2 2021 Hypercalcemia is a rare yet important side-effect of ATRA, especially when it is used concomitantly with a medication that impedes its metabolism by inhibiting cytochrome P-450 in the liver and thus increasing the duration of exposure to ATRA. Tretinoin 53-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. Famotidine 118-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. Famotidine 118-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-243 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. fostemsavir 136-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. Temsavir 156-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. Temsavir 156-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-243 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. fostemsavir 287-298 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. Rifampin 334-342 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. Carbamazepine 344-357 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. Phenytoin 359-368 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. Mitotane 370-378 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33547469-4 2021 As absorption of temsavir is not altered with increased gastric pH, patients may take acid suppressive agents such as famotidine during fostemsavir therapy.Temsavir is primarily metabolized through hydrolysis but also via cytochrome P-450 (CYP) oxidation; therefore, coadministration of fostemsavir with strong CYP3A inducers such as rifampin, carbamazepine, phenytoin, mitotane, enzalutamide, or St John"s wort is contraindicated because it may result in significantly lower temsavir exposure, which can ultimately impair virologic response. enzalutamide 380-392 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-238 33078993-0 2021 Assessment of the in vitro cytochrome P450 (CYP) inhibition potential of Nafithromycin, a next generation lactone ketolide antibiotic. nafithromycin 73-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 33078993-0 2021 Assessment of the in vitro cytochrome P450 (CYP) inhibition potential of Nafithromycin, a next generation lactone ketolide antibiotic. nafithromycin 73-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 33078993-4 2021 To facilitate comparative assessment of drug-drug interaction potential, CYP inhibitory activities of nafithromycin was evaluated in comparison with clarithromycin, telithromycin, cethromycin and solithromycin. nafithromycin 102-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 33747646-4 2021 These medications inhibit two vital enzymes of cytochrome P-450, CYP2C9 and CYP3A4, potentiating the effects of ATRA on calcium metabolism. Tretinoin 112-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 33747646-4 2021 These medications inhibit two vital enzymes of cytochrome P-450, CYP2C9 and CYP3A4, potentiating the effects of ATRA on calcium metabolism. Calcium 120-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 32725383-4 2021 RESULTS: Seventy-two metabolic drug interaction studies were identified where volume of distribution at steady-state (Vss) values were available for the CYP index substrates caffeine (CYP1A2), metoprolol (CYP2D6), midazolam (CYP3A4), theophylline (CYP1A2), and tolbutamide (CYP2C9). Caffeine 174-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-156 32888176-0 2021 Levomepromazine and clozapine induce the main human cytochrome P450 drug metabolizing enzyme CYP3A4. Methotrimeprazine 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 32869328-4 2021 A clinical study in healthy subjects (n = 27) evaluated the inhibition of CYP3A4, CYP2C8, and CYP2C9 in vivo by administering single doses of probe CYP substrates (midazolam, pioglitazone, and tolbutamide) alone and in combination with relacorilant (350 mg). Pioglitazone 175-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 32869328-4 2021 A clinical study in healthy subjects (n = 27) evaluated the inhibition of CYP3A4, CYP2C8, and CYP2C9 in vivo by administering single doses of probe CYP substrates (midazolam, pioglitazone, and tolbutamide) alone and in combination with relacorilant (350 mg). Tolbutamide 193-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 33717590-14 2021 GSEA results showed that high expressed DYNLRB2 and SPTBN1 were enriched in Drug metabolism cytochrome P450, Cardiac muscle contraction, Retinol metabolism. gsea 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 33717590-14 2021 GSEA results showed that high expressed DYNLRB2 and SPTBN1 were enriched in Drug metabolism cytochrome P450, Cardiac muscle contraction, Retinol metabolism. Vitamin A 137-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 33215946-9 2021 EXPERT OPINION: Overall, the CYP enzymes, depending upon the isoform, play a contributory or protective role in hyperoxic lung injury, and are, therefore, ideal candidates for developing drugs that can treat oxygen-mediated lung injury. Oxygen 208-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 32888176-0 2021 Levomepromazine and clozapine induce the main human cytochrome P450 drug metabolizing enzyme CYP3A4. Clozapine 20-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 32888176-1 2021 BACKGROUND: Cytochrome P450 (CYP) enzymes are involved in the metabolism of many important endogenous substrates (steroids, melatonin), drugs and toxic xenobiotics. Steroids 114-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 32888176-1 2021 BACKGROUND: Cytochrome P450 (CYP) enzymes are involved in the metabolism of many important endogenous substrates (steroids, melatonin), drugs and toxic xenobiotics. Melatonin 124-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 33572963-0 2021 Pharmacokinetic Estimation Models-based Approach to Predict Clinical Implications for CYP Induction by Calcitriol in Human Cryopreserved Hepatocytes and HepaRG Cells. Calcitriol 103-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 33259822-2 2021 Quinoneimines formed by cytochrome P450 (CYP)-mediated oxidation of MFA are considered to be causal metabolites of the toxicity and are detoxified by glutathione conjugation. quinoneimines 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 33259822-2 2021 Quinoneimines formed by cytochrome P450 (CYP)-mediated oxidation of MFA are considered to be causal metabolites of the toxicity and are detoxified by glutathione conjugation. quinoneimines 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 33259822-2 2021 Quinoneimines formed by cytochrome P450 (CYP)-mediated oxidation of MFA are considered to be causal metabolites of the toxicity and are detoxified by glutathione conjugation. Mefenamic Acid 68-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 33259822-2 2021 Quinoneimines formed by cytochrome P450 (CYP)-mediated oxidation of MFA are considered to be causal metabolites of the toxicity and are detoxified by glutathione conjugation. Mefenamic Acid 68-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 33259822-2 2021 Quinoneimines formed by cytochrome P450 (CYP)-mediated oxidation of MFA are considered to be causal metabolites of the toxicity and are detoxified by glutathione conjugation. Glutathione 150-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 33259822-2 2021 Quinoneimines formed by cytochrome P450 (CYP)-mediated oxidation of MFA are considered to be causal metabolites of the toxicity and are detoxified by glutathione conjugation. Glutathione 150-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 33146575-5 2021 The formation of 3-OH-NAB was observed after the incubation of NAB with various cytochrome P450 (CYP) isoforms. 3-oh-nab 17-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 33146575-5 2021 The formation of 3-OH-NAB was observed after the incubation of NAB with various cytochrome P450 (CYP) isoforms. 3-oh-nab 17-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 33146575-5 2021 The formation of 3-OH-NAB was observed after the incubation of NAB with various cytochrome P450 (CYP) isoforms. Nabumetone 22-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 33146575-5 2021 The formation of 3-OH-NAB was observed after the incubation of NAB with various cytochrome P450 (CYP) isoforms. Nabumetone 22-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 33146575-10 2021 Further in vitro inhibition experiments showed that multiple non-CYP enzymes are involved in the formation of 6-MNA from 3-OH-NAB. 6-methoxy-2-naphthylacetic acid 110-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 33146575-10 2021 Further in vitro inhibition experiments showed that multiple non-CYP enzymes are involved in the formation of 6-MNA from 3-OH-NAB. 3-oh-nab 121-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 33572963-2 2021 The purpose of this study was to predict the clinical significance of cytochrome P450 (CYP) induction by calcitriol using in vitro human cryopreserved hepatocytes, HepaRG experimental systems, and various pharmacokinetic estimation models. Calcitriol 105-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 33572963-2 2021 The purpose of this study was to predict the clinical significance of cytochrome P450 (CYP) induction by calcitriol using in vitro human cryopreserved hepatocytes, HepaRG experimental systems, and various pharmacokinetic estimation models. Calcitriol 105-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 33572963-7 2021 Therefore, we conclude that CYP induction by calcitriol treatment would not be clinically significant under typical clinical conditions. Calcitriol 45-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 32827665-0 2020 Cytochrome P450-derived linoleic acid metabolites EpOMEs and DiHOMEs: A review of recent studies. Linoleic Acid 24-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 33461459-2 2021 BACKGROUND: Current pharmacokinetic investigations consider reactive oxygen species formed in microsomal reactions as toxic waste products, whereas our works (Manoj et al., 2016) showed that DROS are the reaction mainstay in cytochrome P450 mediated metabolism and that they play significant roles in explaining several unexplained physiologies (Parashar et al., 2018). Oxygen 69-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 225-240 33096358-0 2021 Curcumin ameliorates mercuric chloride-induced liver injury via modulating cytochrome P450 signaling and Nrf2/HO-1 pathway. Curcumin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 33096358-0 2021 Curcumin ameliorates mercuric chloride-induced liver injury via modulating cytochrome P450 signaling and Nrf2/HO-1 pathway. Mercuric Chloride 21-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 33096358-5 2021 Additionally, we also found that curcumin could suppress inflammatory damage, unbalance of trace elements (including sodium, magnesium, kalium, calcium overload), oxidative burst induced by HgCl2, which could be associated with cytochrome P450 (CYP450) signaling. Curcumin 33-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 33096358-5 2021 Additionally, we also found that curcumin could suppress inflammatory damage, unbalance of trace elements (including sodium, magnesium, kalium, calcium overload), oxidative burst induced by HgCl2, which could be associated with cytochrome P450 (CYP450) signaling. Mercuric Chloride 190-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 33411110-1 2022 PURPOSE: Rivaroxaban, an oral anticoagulant, undergoes the metabolism mediated by human cytochrome P450 (CYP). Rivaroxaban 9-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 33411110-1 2022 PURPOSE: Rivaroxaban, an oral anticoagulant, undergoes the metabolism mediated by human cytochrome P450 (CYP). Rivaroxaban 9-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-108 33411110-4 2022 Furthermore, the catalytic efficiency of CYP isoforms was compared via metabolic kinetic studies of rivaroxaban with recombinant CYP isoenzymes, as well as via CYP-specific inhibitory studies. Rivaroxaban 100-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 33411110-6 2022 RESULTS: Multiple CYP isoforms were involved in the hydroxylation of rivaroxaban, with decreasing catalytic rates as follows: CYP2J2 > 3A4 > 2D6 > 4F3 > 1A1 > 3A5 > 3A7 > 2A6 > 2E1 > 2C9 > 2C19. Rivaroxaban 69-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 32979584-0 2021 Interspecies differences in cytochrome P450-mediated metabolism of neonicotinoids among cats, dogs, rats, and humans. Neonicotinoids 67-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 32979584-3 2021 Cytochrome P450 (CYP) is among the most significant xenobiotic metabolizing enzymes that oxidizes several chemicals, including neonicotinoids. Neonicotinoids 127-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32979584-3 2021 Cytochrome P450 (CYP) is among the most significant xenobiotic metabolizing enzymes that oxidizes several chemicals, including neonicotinoids. Neonicotinoids 127-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 32979584-4 2021 However, CYP activities and metabolite compositions of neonicotinoid metabolites are unknown in most domesticated pet species. Neonicotinoids 55-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 32979584-9 2021 CYP activities in metabolism of acetamiprid to dm-acetamiprid and N-acetyl-acetamiprid were determined to be significantly higher in humans compared to other species. acetamiprid 32-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 32979584-9 2021 CYP activities in metabolism of acetamiprid to dm-acetamiprid and N-acetyl-acetamiprid were determined to be significantly higher in humans compared to other species. dm-acetamiprid 47-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 32979584-9 2021 CYP activities in metabolism of acetamiprid to dm-acetamiprid and N-acetyl-acetamiprid were determined to be significantly higher in humans compared to other species. n-acetyl-acetamiprid 66-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 32979584-10 2021 However, further studies should be targeted at identifying the differences in hepatic metabolism of neonicotinoids in these species using recombinant CYP enzymes. Neonicotinoids 100-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 32696500-1 2021 The glycopeptide antibiotics (GPAs) are a fascinating example of complex natural product biosynthesis, with the non-ribosomal synthesis of the peptide core coupled to a cytochrome P450-mediated cyclisation cascade that crosslinks aromatic side chains within this peptide. glycopeptide antibiotics 4-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-184 32696500-1 2021 The glycopeptide antibiotics (GPAs) are a fascinating example of complex natural product biosynthesis, with the non-ribosomal synthesis of the peptide core coupled to a cytochrome P450-mediated cyclisation cascade that crosslinks aromatic side chains within this peptide. gpas 30-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-184 33310663-1 2021 Cytochrome P450 4F (CYP4F) enzymes are responsible for the metabolism of eicosanoids, which play important roles in inflammation. Eicosanoids 73-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-18 33310663-1 2021 Cytochrome P450 4F (CYP4F) enzymes are responsible for the metabolism of eicosanoids, which play important roles in inflammation. Eicosanoids 73-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-25 33388475-0 2021 Regulation of the cytochrome P450 epoxyeicosanoid pathway is associated with distinct histologic features in pediatric non-alcoholic fatty liver disease. epoxyeicosanoid 34-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 33388475-2 2021 Preclinical studies indicate that epoxyeicosanoids, a class of bioactive lipid mediators that are generated by cytochrome P450 (CYP) epoxygenases and inactivated by the soluble epoxide hydrolase (sEH), play a protective role in NAFLD. epoxyeicosanoids 34-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-126 33388475-2 2021 Preclinical studies indicate that epoxyeicosanoids, a class of bioactive lipid mediators that are generated by cytochrome P450 (CYP) epoxygenases and inactivated by the soluble epoxide hydrolase (sEH), play a protective role in NAFLD. epoxyeicosanoids 34-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 33252269-3 2020 The aim of this study was to investigate the relationship between polymorphisms of the drug-metabolizing enzyme gene, cytochrome P450 (CYP450), and the drug transporter gene, ATP-binding cassette subfamily B member 1 (ABCB1), as well as their DNA methylation status with the pathogenesis of steroid-induced ONFH. Steroids 291-298 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 32827665-2 2020 Cytochrome P450-derived LA metabolites 9,10-epoxyoctadecenoic acid (9,10-EpOME), 12,13-epoxyoctadecenoic acid (12,13-EpOME), 9,10-dihydroxy-12Z-octadecenoic acid (9,10-DiHOME) and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) have been studied for their association with various disease states and biological functions. Coronaric acid 39-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32827665-2 2020 Cytochrome P450-derived LA metabolites 9,10-epoxyoctadecenoic acid (9,10-EpOME), 12,13-epoxyoctadecenoic acid (12,13-EpOME), 9,10-dihydroxy-12Z-octadecenoic acid (9,10-DiHOME) and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) have been studied for their association with various disease states and biological functions. 12,13-epoxyoctadecenoic acid 81-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32827665-2 2020 Cytochrome P450-derived LA metabolites 9,10-epoxyoctadecenoic acid (9,10-EpOME), 12,13-epoxyoctadecenoic acid (12,13-EpOME), 9,10-dihydroxy-12Z-octadecenoic acid (9,10-DiHOME) and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) have been studied for their association with various disease states and biological functions. Leukotoxin diol 125-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32827665-2 2020 Cytochrome P450-derived LA metabolites 9,10-epoxyoctadecenoic acid (9,10-EpOME), 12,13-epoxyoctadecenoic acid (12,13-EpOME), 9,10-dihydroxy-12Z-octadecenoic acid (9,10-DiHOME) and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) have been studied for their association with various disease states and biological functions. Leukotoxin diol 163-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32827665-2 2020 Cytochrome P450-derived LA metabolites 9,10-epoxyoctadecenoic acid (9,10-EpOME), 12,13-epoxyoctadecenoic acid (12,13-EpOME), 9,10-dihydroxy-12Z-octadecenoic acid (9,10-DiHOME) and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) have been studied for their association with various disease states and biological functions. (+/-)12,13-Dihome 180-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32827665-2 2020 Cytochrome P450-derived LA metabolites 9,10-epoxyoctadecenoic acid (9,10-EpOME), 12,13-epoxyoctadecenoic acid (12,13-EpOME), 9,10-dihydroxy-12Z-octadecenoic acid (9,10-DiHOME) and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) have been studied for their association with various disease states and biological functions. (+/-)12,13-Dihome 218-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32223485-0 2020 In vitro inhibitory effects of cepharanthine on human liver cytochrome P450 enzymes. cepharanthine 31-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 32673137-0 2020 In vitro inhibitory effect of lysionotin on the activity of cytochrome P450 enzymes. nevadensin 30-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 32673137-2 2020 A study of lysionotin on the activity of human liver cytochrome P450 (CYP) enzymes can provide guidance on the clinical application of lysionotin. nevadensin 11-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 32673137-2 2020 A study of lysionotin on the activity of human liver cytochrome P450 (CYP) enzymes can provide guidance on the clinical application of lysionotin. nevadensin 11-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 32673137-2 2020 A study of lysionotin on the activity of human liver cytochrome P450 (CYP) enzymes can provide guidance on the clinical application of lysionotin. nevadensin 135-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 32673137-2 2020 A study of lysionotin on the activity of human liver cytochrome P450 (CYP) enzymes can provide guidance on the clinical application of lysionotin. nevadensin 135-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 32279279-0 2020 The atypical neuroleptics iloperidone and lurasidone inhibit human cytochrome P450 enzymes in vitro. iloperidone 26-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 32279279-0 2020 The atypical neuroleptics iloperidone and lurasidone inhibit human cytochrome P450 enzymes in vitro. Lurasidone Hydrochloride 42-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 32223485-3 2020 In addition, the enzyme kinetic parameters were calculated.Results: The results showed that the activity of CYP3A4, CYP2E1 and CYP2C9 was inhibited by CEP, with IC50 values of 16.29, 25.62 and 24.57 muM, respectively, but other CYP isoforms were not affected. cepharanthine 151-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 32279279-2 2020 BACKGROUND: The present study aimed at examining the inhibitory effect of two atypical neuroleptics iloperidone and lurasidone on the main human cytochrome P450 (CYP) enzymes in pooled human liver microsomes and cDNA-expressed CYP enzymes (supersomes). iloperidone 100-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 32279279-2 2020 BACKGROUND: The present study aimed at examining the inhibitory effect of two atypical neuroleptics iloperidone and lurasidone on the main human cytochrome P450 (CYP) enzymes in pooled human liver microsomes and cDNA-expressed CYP enzymes (supersomes). iloperidone 100-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 32791187-6 2020 OBJECTIVE: To test the hypothesis that hyperoxia induces greater lung injury and inflammation in Nrf2-/- mice compared to wild type (WT) that differs between sexes, and that this phenotype will be rescued by the administration of the cytochrome P450 (CYP) 1A inducer beta-naphthoflavone (BNF). beta-Naphthoflavone 267-286 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-258 32279279-2 2020 BACKGROUND: The present study aimed at examining the inhibitory effect of two atypical neuroleptics iloperidone and lurasidone on the main human cytochrome P450 (CYP) enzymes in pooled human liver microsomes and cDNA-expressed CYP enzymes (supersomes). iloperidone 100-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 227-230 32279279-2 2020 BACKGROUND: The present study aimed at examining the inhibitory effect of two atypical neuroleptics iloperidone and lurasidone on the main human cytochrome P450 (CYP) enzymes in pooled human liver microsomes and cDNA-expressed CYP enzymes (supersomes). Lurasidone Hydrochloride 116-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 32279279-2 2020 BACKGROUND: The present study aimed at examining the inhibitory effect of two atypical neuroleptics iloperidone and lurasidone on the main human cytochrome P450 (CYP) enzymes in pooled human liver microsomes and cDNA-expressed CYP enzymes (supersomes). Lurasidone Hydrochloride 116-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 32279279-2 2020 BACKGROUND: The present study aimed at examining the inhibitory effect of two atypical neuroleptics iloperidone and lurasidone on the main human cytochrome P450 (CYP) enzymes in pooled human liver microsomes and cDNA-expressed CYP enzymes (supersomes). Lurasidone Hydrochloride 116-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 227-230 32279279-3 2020 METHODS: The activity of these enzymes was determined by the following CYP-specific reactions: caffeine 3-N-demethylation/CYP1A2, diclofenac 4"-hydroxylation/CYP2C9, perazine N-demethylation/CYP2C19, bufuralol 1"-hydroxylation/CYP2D6 and testosterone 6beta-hydroxylation/CYP3A4, respectively, using HPLC. Caffeine 95-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 32279279-3 2020 METHODS: The activity of these enzymes was determined by the following CYP-specific reactions: caffeine 3-N-demethylation/CYP1A2, diclofenac 4"-hydroxylation/CYP2C9, perazine N-demethylation/CYP2C19, bufuralol 1"-hydroxylation/CYP2D6 and testosterone 6beta-hydroxylation/CYP3A4, respectively, using HPLC. Diclofenac 130-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 32279279-3 2020 METHODS: The activity of these enzymes was determined by the following CYP-specific reactions: caffeine 3-N-demethylation/CYP1A2, diclofenac 4"-hydroxylation/CYP2C9, perazine N-demethylation/CYP2C19, bufuralol 1"-hydroxylation/CYP2D6 and testosterone 6beta-hydroxylation/CYP3A4, respectively, using HPLC. Perazine 166-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 32279279-3 2020 METHODS: The activity of these enzymes was determined by the following CYP-specific reactions: caffeine 3-N-demethylation/CYP1A2, diclofenac 4"-hydroxylation/CYP2C9, perazine N-demethylation/CYP2C19, bufuralol 1"-hydroxylation/CYP2D6 and testosterone 6beta-hydroxylation/CYP3A4, respectively, using HPLC. bufuralol 200-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 32279279-3 2020 METHODS: The activity of these enzymes was determined by the following CYP-specific reactions: caffeine 3-N-demethylation/CYP1A2, diclofenac 4"-hydroxylation/CYP2C9, perazine N-demethylation/CYP2C19, bufuralol 1"-hydroxylation/CYP2D6 and testosterone 6beta-hydroxylation/CYP3A4, respectively, using HPLC. Testosterone 238-250 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 32672501-2 2020 Pyrethroid metabolism was also studied with some human expressed cytochrome P450 (CYP) and carboxylesterase (CES) enzymes. Pyrethrins 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 32672501-2 2020 Pyrethroid metabolism was also studied with some human expressed cytochrome P450 (CYP) and carboxylesterase (CES) enzymes. Pyrethrins 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 32672501-10 2020 All pyrethroids were metabolised by some of the human expressed CYP enzymes studied and apart from BIF were also metabolised by CES enzymes. Pyrethrins 4-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 32672501-0 2020 Metabolism of bifenthrin, beta-cyfluthrin, lambda-cyhalothrin, cyphenothrin and esfenvalerate by rat and human cytochrome P450 and carboxylesterase enzymes. bifenthrin 14-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-147 32672501-0 2020 Metabolism of bifenthrin, beta-cyfluthrin, lambda-cyhalothrin, cyphenothrin and esfenvalerate by rat and human cytochrome P450 and carboxylesterase enzymes. cyfluthrin 26-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-147 32672501-0 2020 Metabolism of bifenthrin, beta-cyfluthrin, lambda-cyhalothrin, cyphenothrin and esfenvalerate by rat and human cytochrome P450 and carboxylesterase enzymes. cyhalothrin 43-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-147 32672501-0 2020 Metabolism of bifenthrin, beta-cyfluthrin, lambda-cyhalothrin, cyphenothrin and esfenvalerate by rat and human cytochrome P450 and carboxylesterase enzymes. cyphenothrin 63-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-147 32672501-0 2020 Metabolism of bifenthrin, beta-cyfluthrin, lambda-cyhalothrin, cyphenothrin and esfenvalerate by rat and human cytochrome P450 and carboxylesterase enzymes. fenvalerate 80-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-147 32761661-0 2020 How do metal ions modulate the rate-determining electron transfer step in Cytochrome P450 reactions? Metals 7-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 32761661-3 2020 To understand the effect of external perturbations on the CYP450 first reduction step, we performed a computational study with model complexes in the presence of metal and organic ions, solvent molecules and an electric field effect. Metals 162-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-64 32761661-6 2020 Calculations on a large crystal structure with alkali metal ions bound implicated inhibition patterns of the ions; therefore, we predict that active forms of the natural CYP450 isozymes will not have more than one alkali metal ions bound in the second-coordination sphere. Metals 54-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-176 32761661-6 2020 Calculations on a large crystal structure with alkali metal ions bound implicated inhibition patterns of the ions; therefore, we predict that active forms of the natural CYP450 isozymes will not have more than one alkali metal ions bound in the second-coordination sphere. Metals 221-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-176 33175492-2 2020 By the use of a combinatorial and modular approach for gene expression, including a cytochrome P450 and the corresponding reductase, engineered Synechoccous elongatus PCC 7942 as a model cyanobacterium enabled the biosynthesis of ent-kaurenoic acid at 2.9 +- 0.01 mg L-1 from CO2. kaurenoic acid 230-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 33175492-2 2020 By the use of a combinatorial and modular approach for gene expression, including a cytochrome P450 and the corresponding reductase, engineered Synechoccous elongatus PCC 7942 as a model cyanobacterium enabled the biosynthesis of ent-kaurenoic acid at 2.9 +- 0.01 mg L-1 from CO2. Carbon Dioxide 276-279 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 32791187-6 2020 OBJECTIVE: To test the hypothesis that hyperoxia induces greater lung injury and inflammation in Nrf2-/- mice compared to wild type (WT) that differs between sexes, and that this phenotype will be rescued by the administration of the cytochrome P450 (CYP) 1A inducer beta-naphthoflavone (BNF). beta-Naphthoflavone 288-291 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-258 33201326-3 2022 Among cytochrome P-450 (CYP) isoforms in humans, CYP3A4 is the major isoform involved in metabolic decyclopropylmethylation of nalfurafine hydrochloride. decyclopropylmethylation 99-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 33201326-3 2022 Among cytochrome P-450 (CYP) isoforms in humans, CYP3A4 is the major isoform involved in metabolic decyclopropylmethylation of nalfurafine hydrochloride. decyclopropylmethylation 99-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 33201326-3 2022 Among cytochrome P-450 (CYP) isoforms in humans, CYP3A4 is the major isoform involved in metabolic decyclopropylmethylation of nalfurafine hydrochloride. TRK 820 127-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 33201326-3 2022 Among cytochrome P-450 (CYP) isoforms in humans, CYP3A4 is the major isoform involved in metabolic decyclopropylmethylation of nalfurafine hydrochloride. TRK 820 127-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 32763298-5 2020 Cytochrome P450 (CYP450) and its isoforms are the main metabolizers of HCQ and CQ. Hydroxychloroquine 71-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 33207662-0 2020 Cytochrome P450 Metabolism of Polyunsaturated Fatty Acids and Neurodegeneration. Fatty Acids, Unsaturated 30-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 33207662-4 2020 Recent research demonstrated that the oxidized metabolites, particularly the cytochrome P450 (CYP) metabolites, of PUFAs are beneficial to several neurodegenerative diseases, including Alzheimer"s disease and Parkinson"s disease; however, their mechanism(s) remains unclear. Fatty Acids, Unsaturated 115-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 33207662-4 2020 Recent research demonstrated that the oxidized metabolites, particularly the cytochrome P450 (CYP) metabolites, of PUFAs are beneficial to several neurodegenerative diseases, including Alzheimer"s disease and Parkinson"s disease; however, their mechanism(s) remains unclear. Fatty Acids, Unsaturated 115-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 33207662-8 2020 We will also discuss the potential mechanism(s) of CYP PUFA metabolites in neurodegeneration, which will ultimately improve our understanding of how PUFAs affect neurodegeneration and may identify potential drug targets for neurodegenerative diseases. Fatty Acids, Unsaturated 55-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 32763298-5 2020 Cytochrome P450 (CYP450) and its isoforms are the main metabolizers of HCQ and CQ. Chloroquine 72-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32889110-7 2020 Dip-based CMCs not only alter Dip-mediated inhibition or activation of CYP enzymes but also change the degree to which rCYPs are involved in Dip metabolism. dipfluzine 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 32889110-7 2020 Dip-based CMCs not only alter Dip-mediated inhibition or activation of CYP enzymes but also change the degree to which rCYPs are involved in Dip metabolism. dipfluzine 30-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 32889110-7 2020 Dip-based CMCs not only alter Dip-mediated inhibition or activation of CYP enzymes but also change the degree to which rCYPs are involved in Dip metabolism. dipfluzine 30-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 32889110-10 2020 The effects of the alterations of Dip CMCs on the interaction between Dip and CYP enzymes are not attributed to a simple superposition of Dip and the respective precursor and may be due to the presence of interaction forces between Dip and precursor molecules. dipfluzine 34-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 32889110-10 2020 The effects of the alterations of Dip CMCs on the interaction between Dip and CYP enzymes are not attributed to a simple superposition of Dip and the respective precursor and may be due to the presence of interaction forces between Dip and precursor molecules. dipfluzine 70-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 32889110-10 2020 The effects of the alterations of Dip CMCs on the interaction between Dip and CYP enzymes are not attributed to a simple superposition of Dip and the respective precursor and may be due to the presence of interaction forces between Dip and precursor molecules. dipfluzine 70-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 32889110-11 2020 These results are the first to provide preliminary experimental evidence that CMCs change the interaction between API and CYP enzymes. CMCS 78-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 32941854-0 2020 Asenapine and iloperidone decrease the expression of major cytochrome P450 enzymes CYP1A2 and CYP3A4 in human hepatocytes. asenapine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 33879433-0 2020 Hepato-protective role of itraconazole mediated cytochrome p450 pathway inhibition in liver fibrosis. Itraconazole 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 33879433-6 2020 Based upon this fact, present study is aimed to evaluate the repurposing of Itraconazole in the prevention of hepatic fibrosis via inhibition of cytochrome P450 pathway. Itraconazole 76-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 32941854-0 2020 Asenapine and iloperidone decrease the expression of major cytochrome P450 enzymes CYP1A2 and CYP3A4 in human hepatocytes. iloperidone 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 32941854-4 2020 The present study aimed at examining the effects of the three atypical neuroleptics asenapine, lurasidone and iloperidone on cytochrome P450 (CYP) expression in the human liver. asenapine 84-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-140 32941854-4 2020 The present study aimed at examining the effects of the three atypical neuroleptics asenapine, lurasidone and iloperidone on cytochrome P450 (CYP) expression in the human liver. asenapine 84-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-145 32941854-4 2020 The present study aimed at examining the effects of the three atypical neuroleptics asenapine, lurasidone and iloperidone on cytochrome P450 (CYP) expression in the human liver. Lurasidone Hydrochloride 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-140 32941854-4 2020 The present study aimed at examining the effects of the three atypical neuroleptics asenapine, lurasidone and iloperidone on cytochrome P450 (CYP) expression in the human liver. Lurasidone Hydrochloride 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-145 32941854-4 2020 The present study aimed at examining the effects of the three atypical neuroleptics asenapine, lurasidone and iloperidone on cytochrome P450 (CYP) expression in the human liver. iloperidone 110-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-140 32941854-4 2020 The present study aimed at examining the effects of the three atypical neuroleptics asenapine, lurasidone and iloperidone on cytochrome P450 (CYP) expression in the human liver. iloperidone 110-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-145 32941854-7 2020 CYP activities were measured in the incubation medium using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A1/2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19) and testosterone 6beta-hydroxylation (CYP3A4). Caffeine 88-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 32941854-7 2020 CYP activities were measured in the incubation medium using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A1/2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19) and testosterone 6beta-hydroxylation (CYP3A4). Caffeine 88-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 32941854-7 2020 CYP activities were measured in the incubation medium using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A1/2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19) and testosterone 6beta-hydroxylation (CYP3A4). Diclofenac 127-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 32941854-7 2020 CYP activities were measured in the incubation medium using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A1/2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19) and testosterone 6beta-hydroxylation (CYP3A4). Perazine 165-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 32941854-7 2020 CYP activities were measured in the incubation medium using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A1/2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19) and testosterone 6beta-hydroxylation (CYP3A4). Testosterone 204-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 33023027-0 2020 Cytochrome P450 Can Epoxidize an Oxepin to a Reactive 2,3-Epoxyoxepin Intermediate: Potential Insights into Metabolic Ring-Opening of Benzene. Oxepins 33-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 33050335-3 2020 The results indicated that lipoxygenase and cytochrome P450-derived eicosanoids were elevated (5-HETE, 12-HETE, 20-HETE, and 20-COOH-AA), and there appeared to be no differences in levels measured in eyes with tractional retinal detachments versus those without. Eicosanoids 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 33050335-3 2020 The results indicated that lipoxygenase and cytochrome P450-derived eicosanoids were elevated (5-HETE, 12-HETE, 20-HETE, and 20-COOH-AA), and there appeared to be no differences in levels measured in eyes with tractional retinal detachments versus those without. 5-hydroxy-6,8,11,14-eicosatetraenoic acid 95-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 33050335-3 2020 The results indicated that lipoxygenase and cytochrome P450-derived eicosanoids were elevated (5-HETE, 12-HETE, 20-HETE, and 20-COOH-AA), and there appeared to be no differences in levels measured in eyes with tractional retinal detachments versus those without. 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid 103-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 33095237-1 2020 Arachidonic acid can be metabolized in blood vessels by three primary enzymatic pathways; cyclooxygenase (COX), lipoxygenase (LO), and cytochrome P450 (CYP). Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 33095237-1 2020 Arachidonic acid can be metabolized in blood vessels by three primary enzymatic pathways; cyclooxygenase (COX), lipoxygenase (LO), and cytochrome P450 (CYP). Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-155 33095237-7 2020 CYP enzymes produce two types of eicosanoid products: EDHF vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-HETE. Eicosanoids 33-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 33095237-7 2020 CYP enzymes produce two types of eicosanoid products: EDHF vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-HETE. eets 98-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 33095237-7 2020 CYP enzymes produce two types of eicosanoid products: EDHF vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-HETE. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 128-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 33093609-5 2020 Pathway enrichment analysis revealed that drug metabolism-cytochrome P450, biopterin metabolism, vitamin B9 (folate) metabolism, selenoamino acid metabolism, and methionine and cysteine metabolism showed significant enrichment in vitiligo patients compared with the status in healthy controls. Folic Acid 109-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 33060725-1 2020 Cytochrome P450 (CYP) is involved in the metabolism of nevirapine (NVP); especially, CYP2B6 has been known to be one of the main enzymes involved in NVP metabolism. Nevirapine 55-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 33060725-1 2020 Cytochrome P450 (CYP) is involved in the metabolism of nevirapine (NVP); especially, CYP2B6 has been known to be one of the main enzymes involved in NVP metabolism. Nevirapine 55-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 33023027-0 2020 Cytochrome P450 Can Epoxidize an Oxepin to a Reactive 2,3-Epoxyoxepin Intermediate: Potential Insights into Metabolic Ring-Opening of Benzene. oxepin epoxide 54-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 33023027-0 2020 Cytochrome P450 Can Epoxidize an Oxepin to a Reactive 2,3-Epoxyoxepin Intermediate: Potential Insights into Metabolic Ring-Opening of Benzene. Benzene 134-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 33023027-2 2020 The present study demonstrates that separate incubations of 4,5-benzoxepin with three cytochrome P450 isoforms (2E1, 1A2, and 3A4) as well as pooled human liver microsomes (pHLM) also produce 1H-2-benzopyran-1-carboxaldehyde as the major product, likely via the 2,3-epoxyoxepin. 4,5-benzoxepin 60-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 33023027-2 2020 The present study demonstrates that separate incubations of 4,5-benzoxepin with three cytochrome P450 isoforms (2E1, 1A2, and 3A4) as well as pooled human liver microsomes (pHLM) also produce 1H-2-benzopyran-1-carboxaldehyde as the major product, likely via the 2,3-epoxyoxepin. 1H-isochromene-1-carbaldehyde 192-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 33023027-2 2020 The present study demonstrates that separate incubations of 4,5-benzoxepin with three cytochrome P450 isoforms (2E1, 1A2, and 3A4) as well as pooled human liver microsomes (pHLM) also produce 1H-2-benzopyran-1-carboxaldehyde as the major product, likely via the 2,3-epoxyoxepin. oxepin epoxide 262-277 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 31658882-1 2020 As an important member of cytochrome P450 (CYP) enzymes, human CYP1A2 is associated with the metabolism of caffeine and melatonin and the activation of precarcinogens. Caffeine 107-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 31658882-1 2020 As an important member of cytochrome P450 (CYP) enzymes, human CYP1A2 is associated with the metabolism of caffeine and melatonin and the activation of precarcinogens. Caffeine 107-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 31658882-1 2020 As an important member of cytochrome P450 (CYP) enzymes, human CYP1A2 is associated with the metabolism of caffeine and melatonin and the activation of precarcinogens. Melatonin 120-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 31658882-1 2020 As an important member of cytochrome P450 (CYP) enzymes, human CYP1A2 is associated with the metabolism of caffeine and melatonin and the activation of precarcinogens. Melatonin 120-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 32797061-10 2020 Eicosanoids were grouped by biosynthetic pathway, defined by (1) the fatty acid precursor, including linoleic acid (LA), arachidonic acid (AA), docosahexaenoic acid (DHA), or eicosapentaenoic acid (EPA), and (2) the enzyme group, including cyclooxygenase (COX), lipoxygenase (LOX), or cytochrome P450 (CYP). Eicosanoids 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 285-300 32681429-0 2020 Altered expression of cytochrome P450 enzymes involved in metabolism of androgens and vitamin D in the prostate as a risk factor for prostate cancer. Vitamin D 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 32808658-2 2020 Eicosanoids are oxidised derivatives of 20-carbon polyunsaturated fatty acids (PUFAs) formed by the cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (cytP450) pathways. Eicosanoids 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 32808658-2 2020 Eicosanoids are oxidised derivatives of 20-carbon polyunsaturated fatty acids (PUFAs) formed by the cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (cytP450) pathways. Eicosanoids 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-169 32808658-2 2020 Eicosanoids are oxidised derivatives of 20-carbon polyunsaturated fatty acids (PUFAs) formed by the cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (cytP450) pathways. 20-carbon polyunsaturated fatty acids 40-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 32808658-2 2020 Eicosanoids are oxidised derivatives of 20-carbon polyunsaturated fatty acids (PUFAs) formed by the cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (cytP450) pathways. 20-carbon polyunsaturated fatty acids 40-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-169 32808658-2 2020 Eicosanoids are oxidised derivatives of 20-carbon polyunsaturated fatty acids (PUFAs) formed by the cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (cytP450) pathways. Fatty Acids, Unsaturated 79-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 32808658-2 2020 Eicosanoids are oxidised derivatives of 20-carbon polyunsaturated fatty acids (PUFAs) formed by the cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (cytP450) pathways. Fatty Acids, Unsaturated 79-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-169 32249465-2 2020 It is a pro-drug that is extensively metabolized to carebastine, its active carboxylic acid metabolite, by the hepatic microsomal isoenzyme cytochrome P450 (CYP). carebastine 52-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-155 32249465-2 2020 It is a pro-drug that is extensively metabolized to carebastine, its active carboxylic acid metabolite, by the hepatic microsomal isoenzyme cytochrome P450 (CYP). carebastine 52-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 32249465-2 2020 It is a pro-drug that is extensively metabolized to carebastine, its active carboxylic acid metabolite, by the hepatic microsomal isoenzyme cytochrome P450 (CYP). Carboxylic Acids 76-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-155 32249465-2 2020 It is a pro-drug that is extensively metabolized to carebastine, its active carboxylic acid metabolite, by the hepatic microsomal isoenzyme cytochrome P450 (CYP). Carboxylic Acids 76-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 32797061-17 2020 Eicosanoids derived from AA via the CYP and LOX biosynthetic pathways were positively associated with SGA. Eicosanoids 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 33015460-2 2020 TCDD is considered a carcinogen and has proinflammatory influence on animals and humans, promoting free radicals" formation, and binding with the aryl hydrocarbon receptor (AhR) leads to expression of cytochrome p-450 genes that in turn predisposes to mutations. Polychlorinated Dibenzodioxins 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-217 33005200-3 2020 Recombinant CYP450 supersomes and liver microsomes were used to study the metabolic profiles of kurarinone and its inhibitory actions against cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzymes. kurarinone 96-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 32338045-1 2020 BACKGROUND: The benefit of cytochrome P450 (CYP450) enzyme system metabolized medications, especially clopidogrel, was reported more pronounced in smoking than nonsmoking patients, but limited evidence was available from Asian patients. Clopidogrel 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 32907722-5 2020 Bioavailability of hydrocarbons was evident particularly for high molecular weight PAHs, which also caused significant variations of cellular biomarkers, such as cytochrome P450 metabolization in fish, lysosomal membrane destabilization in mussels, genotoxic effects both in fish and molluscs. Hydrocarbons 19-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. fe-n-c 46-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-245 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. fe-n-c 46-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 247-250 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. fe-n-c 46-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 292-295 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. Heme 80-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-245 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. Heme 80-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 247-250 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. Heme 80-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 292-295 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. fe-n 46-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-245 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. fe-n 46-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 247-250 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. fe-n 46-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 292-295 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. fe-n-c 134-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-245 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. fe-n-c 134-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 247-250 32515070-3 2020 By re-visiting and engineering the well-known Fe-N-C electrocatalyst that has a heme-like Fe-N x active sites, herein, we report that Fe-N-C could not only catalyze drug metabolization but also had inhibition behaviors similar to cytochrome P450 (CYP), endowing it a potential replacement of CYP for preliminary evaluation of massive potential chemicals, drug dosing guide and outcome prediction. fe-n-c 134-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 292-295 32700706-3 2020 The Sn(iv)-metalated COF material Sn-CPF-3 exhibits high photocatalytic efficiency and selectivity in aerobic oxidation of sulfides to produce highly value-added sulfoxides with up to 23 334 turnovers and 648 h-1 turnover frequency under visible light irradiation. Sulfides 123-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-42 32700706-3 2020 The Sn(iv)-metalated COF material Sn-CPF-3 exhibits high photocatalytic efficiency and selectivity in aerobic oxidation of sulfides to produce highly value-added sulfoxides with up to 23 334 turnovers and 648 h-1 turnover frequency under visible light irradiation. Sulfoxides 162-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-42 32797061-10 2020 Eicosanoids were grouped by biosynthetic pathway, defined by (1) the fatty acid precursor, including linoleic acid (LA), arachidonic acid (AA), docosahexaenoic acid (DHA), or eicosapentaenoic acid (EPA), and (2) the enzyme group, including cyclooxygenase (COX), lipoxygenase (LOX), or cytochrome P450 (CYP). Eicosanoids 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 302-305 32874920-7 2020 Primary cultured rat and human hepatocytes were considered to be useful for the evaluation of effects of the benzimidazole compounds on their inducibility and inhibitory activities of cytochrome P450 forms. benzimidazole 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-199 32216303-8 2020 Two examples are chosen to demonstrate the synergy advantage in elucidation of metabolic mechanism of triphenyl phosphate and atrazine catalyzed by CYP, respectively, which shows the interplay between experiments and computations allows gaining greater insight than the isolated methods. triphenyl phosphate 102-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. diosmetin 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. diosmetin 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. acacetin 87-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. acacetin 87-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. Catechin 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. Catechin 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. eriodictyol 110-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. eriodictyol 110-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. capillin 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 32746771-6 2021 From the investigation of CYP inhibitors against MAOB, five CYP inhibitors- Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin have expressed inhibitory action against MAOB without any interference with Akt1 and Akt2. capillin 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 32746771-7 2021 This study mainly represents that Galuteolin and Linarin in the Akt pathway can be perceived for OSCC treatment and other five CYP inhibitors - Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin for the treatment of other diseases and cancers caused by overexpression of MAOB. luteolin-7-glucoside 34-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 32746771-7 2021 This study mainly represents that Galuteolin and Linarin in the Akt pathway can be perceived for OSCC treatment and other five CYP inhibitors - Diosmetin, Acacetin, Epicatechin, Eriodictyol and Capillin for the treatment of other diseases and cancers caused by overexpression of MAOB. linarin 49-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 32778735-5 2020 The results showed DPTM exhibited a moderate inhibitory potential against CYP3A/4 (IC50 values of 10 +- 2 and 8 +- 2 muM, respectively) and weak against the other CYP enzymes based on their IC50 values. dptm 19-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 32778735-6 2020 Compared to the control, CYP isoforms and their transcriptional regulators mRNA expressions significantly increased when the Hep G2 cells were treated with DPTM for a certain period of time. dptm 156-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 32385545-1 2020 PURPOSE: Cytochrome P450 (CYP) is involved in the metabolism of valproic acid (VPA). Valproic Acid 64-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-24 32385545-1 2020 PURPOSE: Cytochrome P450 (CYP) is involved in the metabolism of valproic acid (VPA). Valproic Acid 64-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 32385545-1 2020 PURPOSE: Cytochrome P450 (CYP) is involved in the metabolism of valproic acid (VPA). Valproic Acid 79-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-24 32385545-1 2020 PURPOSE: Cytochrome P450 (CYP) is involved in the metabolism of valproic acid (VPA). Valproic Acid 79-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 32519194-8 2020 The most frequently prescribed CYP inhibitors were melperone (n = 2504, 28.1%) and duloxetine (n = 1324, 14.9%). Duloxetine Hydrochloride 83-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 32519194-10 2020 Carbamazepine was the most frequently prescribed CYP inducer (n = 733, 88.8%). Carbamazepine 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 32519194-14 2020 Physicians should be aware of the CYP inhibitory and inducing potential of psychotropic and internistic drugs (especially, melperone). metylperon 123-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 32216303-8 2020 Two examples are chosen to demonstrate the synergy advantage in elucidation of metabolic mechanism of triphenyl phosphate and atrazine catalyzed by CYP, respectively, which shows the interplay between experiments and computations allows gaining greater insight than the isolated methods. Atrazine 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 32537715-0 2020 Assessment of effects of repeated oral doses of fedratinib on inhibition of cytochrome P450 activities in patients with solid tumors using a cocktail approach. Fedratinib 48-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 32655683-0 2020 Multiple circulating alkaloids and saponins from intravenous Kang-Ai injection inhibit human cytochrome P450 and UDP-glucuronosyltransferase isozymes: potential drug-drug interactions. Alkaloids 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-108 32655683-0 2020 Multiple circulating alkaloids and saponins from intravenous Kang-Ai injection inhibit human cytochrome P450 and UDP-glucuronosyltransferase isozymes: potential drug-drug interactions. Saponins 35-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-108 32655683-0 2020 Multiple circulating alkaloids and saponins from intravenous Kang-Ai injection inhibit human cytochrome P450 and UDP-glucuronosyltransferase isozymes: potential drug-drug interactions. kang-ai 61-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-108 32537715-2 2020 In vitro studies indicated that fedratinib was an inhibitor of several cytochrome P450 (CYP) enzymes. Fedratinib 32-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 32537715-9 2020 These results serve as the basis for dose modifications of these CYP substrate drugs when co-administered with fedratinib. Fedratinib 111-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 32537715-2 2020 In vitro studies indicated that fedratinib was an inhibitor of several cytochrome P450 (CYP) enzymes. Fedratinib 32-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 32537715-3 2020 The primary objective of this study was to evaluate the effects of repeated doses of fedratinib on the activity of CYP2D6, CYP2C19, and CYP3A4 in patients with solid tumors using a CYP probe cocktail. Fedratinib 85-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 32325076-1 2020 OBJECTIVE: To explore the pharmacokinetic interaction between isotretinoin, a cytochrome P-450 (CYP) inducer and potent teratogen, and the etonogestrel contraceptive implant. Isotretinoin 62-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 32415468-0 2020 Effect of Cytochrome P450 and ABCB1 Polymorphisms on Imatinib Pharmacokinetics After Single-Dose Administration to Healthy Subjects. Imatinib Mesylate 53-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 32415468-3 2020 OBJECTIVE: To investigate whether polymorphisms in genes encoding cytochrome P450 (CYP) enzymes and ABCB1 transporter affect imatinib pharmacokinetic parameters. Imatinib Mesylate 125-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 32325076-1 2020 OBJECTIVE: To explore the pharmacokinetic interaction between isotretinoin, a cytochrome P-450 (CYP) inducer and potent teratogen, and the etonogestrel contraceptive implant. Isotretinoin 62-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 32065749-1 2020 Cytochrome P450-dependent metabolism of the anti-HIV drug nevirapine (NVP) to 12-hydroxy-NVP (12-OHNVP) has been implicated in NVP toxicities. Nevirapine 58-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31855101-0 2020 A metabolic pathway for the prodrug nabumetone to the pharmacologically active metabolite, 6-methoxy-2-naphthylacetic acid (6-MNA) by non-cytochrome P450 enzymes. nabumetone 36-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-153 31855101-0 2020 A metabolic pathway for the prodrug nabumetone to the pharmacologically active metabolite, 6-methoxy-2-naphthylacetic acid (6-MNA) by non-cytochrome P450 enzymes. 6-methoxy-2-naphthylacetic acid 91-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-153 31855101-0 2020 A metabolic pathway for the prodrug nabumetone to the pharmacologically active metabolite, 6-methoxy-2-naphthylacetic acid (6-MNA) by non-cytochrome P450 enzymes. 6-methoxy-2-naphthylacetic acid 124-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-153 31855101-10 2020 Further in vitro inhibition studies demonstrated that multiple non-cytochrome P450 enzymes are involved in the formation of 6-MNA. 6-methoxy-2-naphthylacetic acid 124-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 32875445-4 2020 Cytochrome P450 metabolism of specifically long-chain fatty acids forms epoxide metabolites, the epoxy-fatty acids (EpFA). long-chain fatty acids 43-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32875445-4 2020 Cytochrome P450 metabolism of specifically long-chain fatty acids forms epoxide metabolites, the epoxy-fatty acids (EpFA). Epoxy Compounds 72-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32875445-4 2020 Cytochrome P450 metabolism of specifically long-chain fatty acids forms epoxide metabolites, the epoxy-fatty acids (EpFA). epoxy-fatty acids 97-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32875445-4 2020 Cytochrome P450 metabolism of specifically long-chain fatty acids forms epoxide metabolites, the epoxy-fatty acids (EpFA). epfa 116-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32065749-1 2020 Cytochrome P450-dependent metabolism of the anti-HIV drug nevirapine (NVP) to 12-hydroxy-NVP (12-OHNVP) has been implicated in NVP toxicities. Nevirapine 70-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32065749-1 2020 Cytochrome P450-dependent metabolism of the anti-HIV drug nevirapine (NVP) to 12-hydroxy-NVP (12-OHNVP) has been implicated in NVP toxicities. 12-Hydroxynevirapine 78-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32065749-1 2020 Cytochrome P450-dependent metabolism of the anti-HIV drug nevirapine (NVP) to 12-hydroxy-NVP (12-OHNVP) has been implicated in NVP toxicities. 12-ohnvp 94-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32083470-0 2020 Biotransformation Mechanism of Pesticides by Cytochrome P450: A DFT Study on Dieldrin. Dieldrin 77-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 32306288-4 2020 Chloroquine is metabolized by cytochrome P450 and renal clearance is responsible for one third of total clearance. Chloroquine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 32112562-3 2020 Because renal dysfunction induces changes in drug metabolism and protein binding that could alter cytochrome P450 inhibition mechanisms, we hypothesized that renal dysfunction alters the impact of the warfarin-amiodarone DDI. Warfarin 201-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 32112562-3 2020 Because renal dysfunction induces changes in drug metabolism and protein binding that could alter cytochrome P450 inhibition mechanisms, we hypothesized that renal dysfunction alters the impact of the warfarin-amiodarone DDI. Amiodarone 210-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 32327267-6 2020 The risk of drug interaction with dotinurad was expected to be low, because it weakly inhibits metabolic enzymes such as cytochrome P450 (CYP). Dotinurad 34-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-136 32327267-6 2020 The risk of drug interaction with dotinurad was expected to be low, because it weakly inhibits metabolic enzymes such as cytochrome P450 (CYP). Dotinurad 34-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). Perazine 151-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). bufuralol 187-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 32219694-0 2020 In vitro inhibition of human cytochrome P450 enzymes by the novel atypical antipsychotic drug asenapine: a prediction of possible drug-drug interactions. asenapine 94-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 32219694-2 2020 The present study aimed at examining the inhibitory effect of the novel antipsychotic drug asenapine on the main CYP enzymes in human liver. asenapine 91-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). bufuralol 187-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). Caffeine 76-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). Testosterone 228-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). Caffeine 76-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). Nitrogen 86-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). Testosterone 228-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). Nitrogen 86-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 32219694-5 2020 The rates of the CYP-specific reactions were assessed in the absence and presence of asenapine using HPLC. asenapine 85-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). Diclofenac 113-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 32219694-4 2020 Activities of CYP enzymes were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6), and testosterone 6beta-hydroxylation (CYP3A4). Diclofenac 113-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 32486438-3 2020 In order to study a possible role for CYP induction in neurorepair, we designed an in vitro model where undifferentiated neuroblastoma SH-SY5Y cells were treated with the CYP inducers beta-naphthoflavone (betaNF) and ethanol (EtOH) before and during exposure to the parkinsonian neurotoxin, MPP+. beta-Naphthoflavone 184-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-174 32483226-0 2020 Green analytical method for the simultaneous analysis of cytochrome P450 probe substrates by poly(N-isopropylacrylamide)-based temperature-responsive chromatography. poly-N-isopropylacrylamide 93-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 32483226-6 2020 This study sought to develop the simultaneous analysis of multiple CYP substrates by using poly(N-isopropylacrylamide) (PNIPAAm)-based temperature-responsive chromatography with only aqueous solvents and isocratic methods. poly-N-isopropylacrylamide 91-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 32483226-6 2020 This study sought to develop the simultaneous analysis of multiple CYP substrates by using poly(N-isopropylacrylamide) (PNIPAAm)-based temperature-responsive chromatography with only aqueous solvents and isocratic methods. poly-N-isopropylacrylamide 120-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 32483226-7 2020 Good separation of multiple CYP substrates was achieved without using organic solvents and any gradient methods by temperature-responsive chromatography utilizing a P(NIPAAm-co-n-butyl methacrylate (BMA))- and P(NIPAAm-co-N-acryloyl L-tryptophan methyl ester (L-Trp-OMe))-grafted silica column. Phosphorus 165-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 32483226-7 2020 Good separation of multiple CYP substrates was achieved without using organic solvents and any gradient methods by temperature-responsive chromatography utilizing a P(NIPAAm-co-n-butyl methacrylate (BMA))- and P(NIPAAm-co-N-acryloyl L-tryptophan methyl ester (L-Trp-OMe))-grafted silica column. nipaam-co-n-butyl methacrylate 167-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 32483226-7 2020 Good separation of multiple CYP substrates was achieved without using organic solvents and any gradient methods by temperature-responsive chromatography utilizing a P(NIPAAm-co-n-butyl methacrylate (BMA))- and P(NIPAAm-co-N-acryloyl L-tryptophan methyl ester (L-Trp-OMe))-grafted silica column. bma 199-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 32483226-7 2020 Good separation of multiple CYP substrates was achieved without using organic solvents and any gradient methods by temperature-responsive chromatography utilizing a P(NIPAAm-co-n-butyl methacrylate (BMA))- and P(NIPAAm-co-N-acryloyl L-tryptophan methyl ester (L-Trp-OMe))-grafted silica column. p(nipaam-co-n-acryloyl l-tryptophan methyl ester 210-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 32483226-7 2020 Good separation of multiple CYP substrates was achieved without using organic solvents and any gradient methods by temperature-responsive chromatography utilizing a P(NIPAAm-co-n-butyl methacrylate (BMA))- and P(NIPAAm-co-N-acryloyl L-tryptophan methyl ester (L-Trp-OMe))-grafted silica column. l-trp-ome 260-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 32483226-7 2020 Good separation of multiple CYP substrates was achieved without using organic solvents and any gradient methods by temperature-responsive chromatography utilizing a P(NIPAAm-co-n-butyl methacrylate (BMA))- and P(NIPAAm-co-N-acryloyl L-tryptophan methyl ester (L-Trp-OMe))-grafted silica column. Silicon Dioxide 280-286 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 32483226-8 2020 Overall, PNIPAAm-based temperature-responsive chromatography represents a remarkably simple, versatile, and environmentally friendly bioanalytical method for CYP substrates and their metabolites. poly-N-isopropylacrylamide 9-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-161 32486438-6 2020 These treatments also protected complex I activity against the inhibitory effects caused by MPP+, suggesting a possible neuroprotective role for CYP inducers. mangion-purified polysaccharide (Candida albicans) 92-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 32201191-0 2020 Identification and in silico prediction of metabolites of tebufenozide derivatives by major human cytochrome P450 isoforms. tebufenozide 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 31919943-1 2020 The regio- (and stereo-) selectivity and specific activity of Cytochrome P450s are determined by the accessibility of potential sites-of-metabolism (SOMs) of the bound substrate relative to the heme, and the activation barrier of the regioselective oxidation reaction. Heme 194-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 32201191-5 2020 In our previous study, we identified the metabolites of tebufenozide, an insect growth regulator, formed by two human CYP isoforms: CYP3A4 and CYP2C19. tebufenozide 56-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 32201191-6 2020 The accessibility of each site of tebufenozide to the reaction center of CYP enzymes and the susceptibility of each hydrogen atom for metabolism by CYP enzymes were evaluated by a docking simulation and hydrogen atom abstraction energy estimation at the density functional theory level, respectively. tebufenozide 34-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 32201191-6 2020 The accessibility of each site of tebufenozide to the reaction center of CYP enzymes and the susceptibility of each hydrogen atom for metabolism by CYP enzymes were evaluated by a docking simulation and hydrogen atom abstraction energy estimation at the density functional theory level, respectively. Hydrogen 116-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 32201191-6 2020 The accessibility of each site of tebufenozide to the reaction center of CYP enzymes and the susceptibility of each hydrogen atom for metabolism by CYP enzymes were evaluated by a docking simulation and hydrogen atom abstraction energy estimation at the density functional theory level, respectively. Hydrogen 203-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 32201191-6 2020 The accessibility of each site of tebufenozide to the reaction center of CYP enzymes and the susceptibility of each hydrogen atom for metabolism by CYP enzymes were evaluated by a docking simulation and hydrogen atom abstraction energy estimation at the density functional theory level, respectively. Hydrogen 203-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 32198084-1 2020 Cytochrome P450 (P450) 2E1 is the major P450 enzyme involved in ethanol metabolism. Ethanol 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 32364297-2 2020 To investigate herb-drug interaction potential associated with deoxyshikonin, we examined the inhibitory effects of deoxyshikonin on eight major cytochrome P450 (CYP) enzymes using cocktail substrate-incubated human liver microsomes. deoxyshikonin 116-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 32406758-3 2020 CYP2C9 is one of the most abundant CYP enzymes and responsible for the metabolism of over 15% clinical drugs, including oral sulfonylurea hypoglycemics, nonsteroidal anti-inflammatory agents, selective cyclooxygenase-2 inhibitors, antiepileptics, angiotensin II receptor inhibitors and anticoagulants. Sulfonylurea Compounds 125-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 31938857-2 2020 Phenytoin is a commonly used drug, and its metabolism is mediated by a specific cytochrome-P450 isoform, CYP2C9, which is encoded by a polymorphic gene. Phenytoin 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 32027168-1 2020 BACKGROUND AND OBJECTIVES: Clopidogrel is widely used after the percutaneous coronary intervention (PCI) in patients with acute coronary syndrome (ACS) and requires activation by cytochrome P450 (CYP), primarily CYP2C19. clopidogrel 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-194 32027168-1 2020 BACKGROUND AND OBJECTIVES: Clopidogrel is widely used after the percutaneous coronary intervention (PCI) in patients with acute coronary syndrome (ACS) and requires activation by cytochrome P450 (CYP), primarily CYP2C19. clopidogrel 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 196-199 32041058-6 2020 Interestingly, ~50% of esterase/lipase/cytochrome P450 genes enriched by DEHP under aerobic conditions were from Nocardioides, a bacterial genus that has not been previously directly linked to phthalate ester degradation. phthalic acid 193-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 32364297-2 2020 To investigate herb-drug interaction potential associated with deoxyshikonin, we examined the inhibitory effects of deoxyshikonin on eight major cytochrome P450 (CYP) enzymes using cocktail substrate-incubated human liver microsomes. deoxyshikonin 116-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 32196986-9 2020 Ataluren showed no or little potential to inhibit or induce most of the CYP enzymes. ataluren 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 32333310-1 2020 Cytochrome p450-mediated metabolism of GRS (indolinone antiaggregant) and its effects on activities of cytochrome p450 isoenzymes were studied. Oxindoles 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 32333310-1 2020 Cytochrome p450-mediated metabolism of GRS (indolinone antiaggregant) and its effects on activities of cytochrome p450 isoenzymes were studied. Oxindoles 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 31628422-2 2020 Cytochrome P450 (CYP) CYP2B6 G516T (rs3745274) is a well-known predictor of efavirenz disposition. efavirenz 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31628422-2 2020 Cytochrome P450 (CYP) CYP2B6 G516T (rs3745274) is a well-known predictor of efavirenz disposition. efavirenz 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 32155912-0 2020 Testing the Pharmacokinetic Interactions of 24 Colonic Flavonoid Metabolites with Human Serum Albumin and Cytochrome P450 Enzymes. Flavonoids 55-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 31733338-0 2020 Cytochrome P450-Derived Eicosanoids and Inflammation in Liver Diseases. Eicosanoids 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31733338-5 2020 Members of omega-3 as well as omega-6 polyunsaturated fatty acids (PUFAs) such as eicosapentaenoic acid and arachidonic acid, respectively, can be metabolized by CYP isoforms leading to the production of biologically active lipid mediators called eicosanoids. Fatty Acids, Unsaturated 30-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 31733338-5 2020 Members of omega-3 as well as omega-6 polyunsaturated fatty acids (PUFAs) such as eicosapentaenoic acid and arachidonic acid, respectively, can be metabolized by CYP isoforms leading to the production of biologically active lipid mediators called eicosanoids. Eicosapentaenoic Acid 82-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 31733338-5 2020 Members of omega-3 as well as omega-6 polyunsaturated fatty acids (PUFAs) such as eicosapentaenoic acid and arachidonic acid, respectively, can be metabolized by CYP isoforms leading to the production of biologically active lipid mediators called eicosanoids. Arachidonic Acids 108-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 31733338-5 2020 Members of omega-3 as well as omega-6 polyunsaturated fatty acids (PUFAs) such as eicosapentaenoic acid and arachidonic acid, respectively, can be metabolized by CYP isoforms leading to the production of biologically active lipid mediators called eicosanoids. Eicosanoids 247-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 31733338-6 2020 CYP-derived eicosanoids have been shown to play significant roles in the pathophysiology and protection of multiple inflammatory liver diseases. Eicosanoids 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 31733338-7 2020 In this review, we elucidate the intricate role of CYP-derived eicosanoids in inflammation in liver diseases paving the way for better therapeutic approaches. Eicosanoids 63-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 31698143-0 2020 Prostaglandins and Other Lipid Mediators Cytochrome P450 derived epoxidized fatty acids as a therapeutic tool against neuroinflammatory diseases. Prostaglandins 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 31698143-0 2020 Prostaglandins and Other Lipid Mediators Cytochrome P450 derived epoxidized fatty acids as a therapeutic tool against neuroinflammatory diseases. Fatty Acids 76-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. Arachidonic Acids 36-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. Arachidonic Acids 36-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. Arachidonic Acids 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. Arachidonic Acids 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. Fatty Acids 68-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. Fatty Acids 68-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. Fatty Acids 87-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. Fatty Acids 87-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. 5,6-epoxy-8,11,14-eicosatrienoic acid 102-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. 5,6-epoxy-8,11,14-eicosatrienoic acid 102-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. 5,6-epoxy-8,11,14-eicosatrienoic acid 129-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31698143-1 2020 Cytochrome P450 (CYP) metabolism of arachidonic acid (ARA) produces epoxy fatty acids (EpFAs) such as epoxyeicosatrienoic acids (EETs) that are known to exert protective effects in inflammatory disorders. 5,6-epoxy-8,11,14-eicosatrienoic acid 129-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 32155912-5 2020 Therefore, in this in vitro study, we investigated the interactions of 24 microbial flavonoid metabolites with human serum albumin and cytochrome P450 (CYP2C9, 2C19, and 3A4) enzymes. Flavonoids 84-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 32131538-0 2020 Mertansine Inhibits mRNA Expression and Enzyme Activities of Cytochrome P450s and Uridine 5"-Diphospho-Glucuronosyltransferases in Human Hepatocytes and Liver Microsomes. Maytansine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 31702786-3 2020 BD undergoes cytochrome P450-mediated metabolic activation to 3,4-epoxy-1-butene (EB), 1,2,3,4-diepoxybutane (DEB) and 1,2-dihydroxy-3,4-epoxybutane (EBD), which form covalent adducts with DNA. 3,4-epoxy-1-butene 62-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 32129155-0 2020 Samarium enriches antitumor activity of ZnO nanoparticles via downregulation of CXCR4 receptor and cytochrome P450. Samarium 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-114 32129155-0 2020 Samarium enriches antitumor activity of ZnO nanoparticles via downregulation of CXCR4 receptor and cytochrome P450. Zinc Oxide 40-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-114 32129155-2 2020 Zinc oxide nanoparticles play an effective role in tumor treatment but with some cautions, such as overexpression of cytochrome P450, hepatic overload, and the mammalian target of rapamycin pathway resistance. Zinc Oxide 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-132 32129155-8 2020 Moreover, samarium:zinc oxide nanoparticles exhibited minimum toxicity which was indicated by suppressed activities of cytochrome P450 and hepatic enzymes, including alanine transaminase and aspartate transaminase. Samarium 10-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-134 32129155-8 2020 Moreover, samarium:zinc oxide nanoparticles exhibited minimum toxicity which was indicated by suppressed activities of cytochrome P450 and hepatic enzymes, including alanine transaminase and aspartate transaminase. Zinc Oxide 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-134 31702786-3 2020 BD undergoes cytochrome P450-mediated metabolic activation to 3,4-epoxy-1-butene (EB), 1,2,3,4-diepoxybutane (DEB) and 1,2-dihydroxy-3,4-epoxybutane (EBD), which form covalent adducts with DNA. 3,4-epoxy-1-butene 82-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 31702786-3 2020 BD undergoes cytochrome P450-mediated metabolic activation to 3,4-epoxy-1-butene (EB), 1,2,3,4-diepoxybutane (DEB) and 1,2-dihydroxy-3,4-epoxybutane (EBD), which form covalent adducts with DNA. diepoxybutane 87-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 31702786-3 2020 BD undergoes cytochrome P450-mediated metabolic activation to 3,4-epoxy-1-butene (EB), 1,2,3,4-diepoxybutane (DEB) and 1,2-dihydroxy-3,4-epoxybutane (EBD), which form covalent adducts with DNA. diepoxybutane 110-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 31702786-3 2020 BD undergoes cytochrome P450-mediated metabolic activation to 3,4-epoxy-1-butene (EB), 1,2,3,4-diepoxybutane (DEB) and 1,2-dihydroxy-3,4-epoxybutane (EBD), which form covalent adducts with DNA. 3,4-epoxybutane-1,2-diol 119-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 31702786-3 2020 BD undergoes cytochrome P450-mediated metabolic activation to 3,4-epoxy-1-butene (EB), 1,2,3,4-diepoxybutane (DEB) and 1,2-dihydroxy-3,4-epoxybutane (EBD), which form covalent adducts with DNA. 3,4-epoxybutane-1,2-diol 150-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 31744669-0 2020 Validation of a sensitive UHPLC-MS/MS method for cytochrome P450 probe substrates caffeine, tolbutamide, dextromethorphan, and alprazolam in human serum reveals drug contamination of serum used for research. Caffeine 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 31744669-0 2020 Validation of a sensitive UHPLC-MS/MS method for cytochrome P450 probe substrates caffeine, tolbutamide, dextromethorphan, and alprazolam in human serum reveals drug contamination of serum used for research. Tolbutamide 92-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 31744669-0 2020 Validation of a sensitive UHPLC-MS/MS method for cytochrome P450 probe substrates caffeine, tolbutamide, dextromethorphan, and alprazolam in human serum reveals drug contamination of serum used for research. Dextromethorphan 105-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 31744669-0 2020 Validation of a sensitive UHPLC-MS/MS method for cytochrome P450 probe substrates caffeine, tolbutamide, dextromethorphan, and alprazolam in human serum reveals drug contamination of serum used for research. Alprazolam 127-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 31679131-6 2020 Etravirine exhibits the potential for bi-directional drug-drug interactions with other antiretrovirals and concomitant medications through its interactions with cytochrome P450 (CYP) isozymes: CYP3A4, CYP2C9, and CYP2C19. etravirine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-176 31092058-5 2020 All three were found to have elevated neuronal and glial auto-antibodies, biomarkers of central nervous system injury, and all three had genetic polymorphisms of paraoxonase (PON-1) and two of cytochrome P450, leading to a reduced ability to metabolize organophosphate compound (OPs).Discussion: A similar constellation of symptoms has been described in other studies of aircrew, although objective evidence of exposure is lacking in most of these studies. Organophosphates 253-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-208 31679131-6 2020 Etravirine exhibits the potential for bi-directional drug-drug interactions with other antiretrovirals and concomitant medications through its interactions with cytochrome P450 (CYP) isozymes: CYP3A4, CYP2C9, and CYP2C19. etravirine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 178-181 31902468-7 2020 In this review, we present important, recent developments in the computational prediction of the inhibition of four clinically crucial CYP isoforms (CYP1A2, 2C9, 2D6, and 3A4) and three nuclear receptors (aryl hydrocarbon receptor, constitutive androstane receptor, and pregnane X receptor) involved in the induction of CYP1A2, 2B6, and 3A4, respectively. Hydrocarbons 205-221 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 31727673-8 2020 Subsequently, oxycodone was metabolized to other metabolites including noroxycodone, noroxymorphone and oxymorphone via cytochrome P-450. Oxycodone 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 31902468-7 2020 In this review, we present important, recent developments in the computational prediction of the inhibition of four clinically crucial CYP isoforms (CYP1A2, 2C9, 2D6, and 3A4) and three nuclear receptors (aryl hydrocarbon receptor, constitutive androstane receptor, and pregnane X receptor) involved in the induction of CYP1A2, 2B6, and 3A4, respectively. androstane 245-255 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 31727673-8 2020 Subsequently, oxycodone was metabolized to other metabolites including noroxycodone, noroxymorphone and oxymorphone via cytochrome P-450. noroxymorphone 85-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 31902468-7 2020 In this review, we present important, recent developments in the computational prediction of the inhibition of four clinically crucial CYP isoforms (CYP1A2, 2C9, 2D6, and 3A4) and three nuclear receptors (aryl hydrocarbon receptor, constitutive androstane receptor, and pregnane X receptor) involved in the induction of CYP1A2, 2B6, and 3A4, respectively. Pregnanes 270-278 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 31629065-7 2020 The cytotoxicity of ADQ was increased in CYP2C8 and 3A4 overexpressing HepG2 cells compared to HepG2/CYP vector cells, confirming that NADQ was more toxic than ADQ. Amodiaquine 20-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 31837525-0 2020 Cytochrome P450 and flavin-containing monooxygenase enzymes are responsible for differential oxidation of the anti-thyroid-cancer drug vandetanib by human and rat hepatic microsomal systems. vandetanib 135-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31494843-14 2020 The in vitro CYP inhibition study only found very weak action for CYP3A4 (midazolam 1"-hydroxylation) and CYP3A4 (midazolam 6beta-hydroxylation) with IC50 of 56.8 microM and 41.1 microM, respectively. Midazolam 74-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 31494843-14 2020 The in vitro CYP inhibition study only found very weak action for CYP3A4 (midazolam 1"-hydroxylation) and CYP3A4 (midazolam 6beta-hydroxylation) with IC50 of 56.8 microM and 41.1 microM, respectively. Midazolam 114-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 31842058-3 2020 Recent research has shown that Cytochrome P450 (CYP) enzymes affect venlafaxine efficacy by mediating its metabolism. venlafaxine 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 31842058-3 2020 Recent research has shown that Cytochrome P450 (CYP) enzymes affect venlafaxine efficacy by mediating its metabolism. venlafaxine 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 31842058-4 2020 The present study investigates genetic polymorphisms of cytochrome P450 family 2 subfamily C member 19 (CYP2C19) are associated with remission after venlafaxine treatment for MDD. venlafaxine 149-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 31859010-4 2020 Further investigation indicated that compound 16h potently suppressed the migration of A549 cells, had moderate stability in rat liver microsomes and showed moderate inhibitory activity against various subtypes of human cytochrome P450. N-[(1R,3R)-3-hydroxy-1-(hydroxymethyl)-3-phenylpropyl]hexadecanamide 46-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 220-235 31979355-4 2020 Tofogliflozin is metabolized and inactivated by five different enzymes CYP2C18, CYP3A4, CYP3A5, CYP4A11, and CYP4F3. 6-((4-ethylphenyl)methyl)-3',4',5',6'-tetrahydro-6'-(hydroxymethyl)spiro(isobenzofuran-1(3H),2'-(2H)pyran)-3',4',5'-triol 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-115 31020909-0 2020 In vitro inhibitory effects of phellodendrine on human liver cytochrome P450 enzymes. phellodendrine 31-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 31020909-3 2020 However, whether phellodendrine affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear.2. phellodendrine 17-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 31020909-3 2020 However, whether phellodendrine affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear.2. phellodendrine 17-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 31020909-4 2020 In this study, the inhibitory effects of phellodendrine on eight human liver CYP isoforms (i.e. 1A2, 3A4, 2A6, 2E1, 2D6, 2C9, 2C19 and 2C8) were investigated in vitro using human liver microsomes (HLMs).3. phellodendrine 41-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 31020909-5 2020 The results showed that phellodendrine inhibited the activity of CYP1A2, 3A4 and 2C9, with IC50 values of 20.56, 14.98 and 16.30 muM, respectively, but that other CYP isoforms were not affected. phellodendrine 24-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 31020909-8 2020 The in vitro studies of phellodendrine with CYP isoforms indicate that phellodendrine could inhibit the activities of CYP1A2, 3A4 and 2C9. phellodendrine 24-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 31020909-8 2020 The in vitro studies of phellodendrine with CYP isoforms indicate that phellodendrine could inhibit the activities of CYP1A2, 3A4 and 2C9. phellodendrine 71-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 31815452-4 2020 (Driscoll, et al 2018) Here cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) reaction phenotyping experiments using recombinant enzymes were performed on bromfenac and BI to identify the CYP and UGT enzymes responsible for bromfenac"s metabolism and bioactivation. bromfenac 165-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 31773494-0 2020 Correction to: Analysis of glutathione S-transferase and cytochrome P450 gene polymorphism in recipients of dose-adjusted busulfan-cyclophosphamide conditioning. Cyclophosphamide 122-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 31648049-11 2020 CYP2C9 and CYP2C19 were the two CYP isozymes that produced 3"-hydroxylnalbuphine and 4"-hydroxylnalbuphine. AN 3 59-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 31648049-11 2020 CYP2C9 and CYP2C19 were the two CYP isozymes that produced 3"-hydroxylnalbuphine and 4"-hydroxylnalbuphine. BE 4-4-4-4 85-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 31555969-0 2020 Analysis of glutathione S-transferase and cytochrome P450 gene polymorphism in recipients of dose-adjusted busulfan-cyclophosphamide conditioning. Cyclophosphamide 107-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 32094037-2 2020 CASE SUMMARY: Polycyclic aromatic hydrocarbons present in cigarette smoke are well-known inducers of cytochrome P450 (CYP) enzymes. Polycyclic Aromatic Hydrocarbons 14-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 32094037-2 2020 CASE SUMMARY: Polycyclic aromatic hydrocarbons present in cigarette smoke are well-known inducers of cytochrome P450 (CYP) enzymes. Polycyclic Aromatic Hydrocarbons 14-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 33327871-9 2020 Based on GO and KEGG analyses, we found that miR-205-5p was correlated with the progression of HNSCC through association with signaling pathways, including the drug metabolism-cytochrome P450 pathway. mir-205-5p 45-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-191 31378969-1 2020 The aim of this study was to characterize the effects of upadacitinib, a Janus kinase 1 inhibitor, on in vivo activity of different cytochrome P450 (CYP) enzymes using a cocktail approach. upadacitinib 57-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-147 31378969-1 2020 The aim of this study was to characterize the effects of upadacitinib, a Janus kinase 1 inhibitor, on in vivo activity of different cytochrome P450 (CYP) enzymes using a cocktail approach. upadacitinib 57-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-152 31956764-1 2020 Cytochrome P450 (CYP450) enzymes belong to a superfamily of heme-containing proteins that are involved in the metabolism of structurally diverse endogenous and exogenous compounds. Heme 60-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31956764-1 2020 Cytochrome P450 (CYP450) enzymes belong to a superfamily of heme-containing proteins that are involved in the metabolism of structurally diverse endogenous and exogenous compounds. Heme 60-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 31730751-3 2019 In this work, we report on the metabolism of BACs in human liver microsomes (HLM) and by recombinant human hepatic cytochrome P450 (CYP) enzymes. Benzalkonium Compounds 45-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-130 31730751-3 2019 In this work, we report on the metabolism of BACs in human liver microsomes (HLM) and by recombinant human hepatic cytochrome P450 (CYP) enzymes. Benzalkonium Compounds 45-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 31730751-12 2019 Furthermore, we demonstrate that CYP-mediated oxidation of C10-BAC mitigates the potent inhibition of cholesterol biosynthesis exhibited by this short-chain BAC. Cholesterol 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 31730751-13 2019 Furthermore, we demonstrate that CYP-mediated oxidation of C10-BAC mitigates the potent inhibition of cholesterol biosynthesis exhibited by this short-chain BAC. Cholesterol 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 31832203-2 2019 The present investigation determined whether DMF and its main metabolite monomethylfumarate (MMF) interact with hepatic cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) transporter, and was performed as part of DMF"s regulatory commitments. Dimethyl Fumarate 45-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 31832203-2 2019 The present investigation determined whether DMF and its main metabolite monomethylfumarate (MMF) interact with hepatic cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) transporter, and was performed as part of DMF"s regulatory commitments. Dimethyl Fumarate 45-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 31832203-4 2019 In vitro inhibition experiments using CYP-selective substrates with human liver microsomes showed 50% inhibitory concentrations (IC50) of >666 micromol/L for DMF and >750 micromol/L for MMF. Dimethyl Fumarate 158-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 31832203-2 2019 The present investigation determined whether DMF and its main metabolite monomethylfumarate (MMF) interact with hepatic cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) transporter, and was performed as part of DMF"s regulatory commitments. citraconic acid 73-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 31832203-4 2019 In vitro inhibition experiments using CYP-selective substrates with human liver microsomes showed 50% inhibitory concentrations (IC50) of >666 micromol/L for DMF and >750 micromol/L for MMF. mmf 186-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 31832203-2 2019 The present investigation determined whether DMF and its main metabolite monomethylfumarate (MMF) interact with hepatic cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) transporter, and was performed as part of DMF"s regulatory commitments. citraconic acid 73-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 31832203-2 2019 The present investigation determined whether DMF and its main metabolite monomethylfumarate (MMF) interact with hepatic cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) transporter, and was performed as part of DMF"s regulatory commitments. mmf 93-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 31832203-2 2019 The present investigation determined whether DMF and its main metabolite monomethylfumarate (MMF) interact with hepatic cytochrome P450 (CYP) enzymes and the P-glycoprotein (P-gp) transporter, and was performed as part of DMF"s regulatory commitments. mmf 93-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 31686406-1 2019 INTRODUCTION: Factor Xa-inhibiting direct oral anticoagulants (FXa-DOACs) undergo hepatic metabolism via cytochrome P-450 (CYP450). doacs 67-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 31605674-9 2019 Cytochrome P450 isozyme models accurately predicted the likelihood for terbinafine N-demethylation, but overestimated the likelihood for a minor N-denaphthylation pathway. terbinafine 71-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31605674-9 2019 Cytochrome P450 isozyme models accurately predicted the likelihood for terbinafine N-demethylation, but overestimated the likelihood for a minor N-denaphthylation pathway. Nitrogen 83-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31605674-9 2019 Cytochrome P450 isozyme models accurately predicted the likelihood for terbinafine N-demethylation, but overestimated the likelihood for a minor N-denaphthylation pathway. Nitrogen 145-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31686406-1 2019 INTRODUCTION: Factor Xa-inhibiting direct oral anticoagulants (FXa-DOACs) undergo hepatic metabolism via cytochrome P-450 (CYP450). doacs 67-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-129 31571146-7 2019 The enzymatic activity of nine CYP isoforms was not inhibited or slightly inhibited in vitro with darolutamide, and a rank order and mechanistic static assessment indicated that risk of clinically relevant DDIs via CYP inhibition is very low. darolutamide 98-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 31571146-13 2019 CONCLUSIONS: Darolutamide has a low potential for clinically relevant DDIs with drugs that are substrates for CYP or P-gp; increased exposure of BCRP and probably OATP substrates was the main interaction of note. darolutamide 13-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 31571146-3 2019 METHODS: In vitro studies were performed to determine the potential for darolutamide to be a substrate, inducer or inhibitor for cytochrome P450 (CYP) isoforms, other metabolizing enzymes and drug transporters. darolutamide 72-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-144 31571146-3 2019 METHODS: In vitro studies were performed to determine the potential for darolutamide to be a substrate, inducer or inhibitor for cytochrome P450 (CYP) isoforms, other metabolizing enzymes and drug transporters. darolutamide 72-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 31172535-0 2019 Drug-Drug Interaction Study to Assess the Effect of Cytochrome P450 Inhibition and Induction on the Pharmacokinetics of the Novel Cereblon Modulator Avadomide (CC-122) in Healthy Adult Subjects. 3-(5-amino-2-methyl-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione 149-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 31172535-0 2019 Drug-Drug Interaction Study to Assess the Effect of Cytochrome P450 Inhibition and Induction on the Pharmacokinetics of the Novel Cereblon Modulator Avadomide (CC-122) in Healthy Adult Subjects. 3-(5-amino-2-methyl-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione 160-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 31456483-0 2019 In vitro inhibitory effects of kaempferitrin on human liver cytochrome P450 enzymes. lespenefril 31-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 31456483-4 2019 Materials and methods: In vitro, HLMs were used to investigate the inhibitory effects of KF (100 muM) on the eight human liver CYP isoforms (i.e., 1A2, 3A4, 2A6, 2E1, 2D6, 2C9, 2C19, and 2C8), and corresponding probe substrates were used. kf 89-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 31456483-3 2019 Objective: This study investigates the effects of KF on eight major CYP isoforms in human liver microsomes (HLMs). kf 50-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 31663733-1 2019 Sterol 14alpha-demethylases (CYP51) are the cytochrome P450 enzymes required for biosynthesis of sterols in eukaryotes, the major targets for antifungal agents and prospective targets for treatment of protozoan infections. Sterols 0-6 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 30865484-8 2019 Using human recombinant CYP450 enzymes and chemical inhibitor method, the metabolism of safrole RMs was predominantly carried out through the CYP1A2 with minor contributions by CYP2E1. Safrole 88-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-30 31663733-1 2019 Sterol 14alpha-demethylases (CYP51) are the cytochrome P450 enzymes required for biosynthesis of sterols in eukaryotes, the major targets for antifungal agents and prospective targets for treatment of protozoan infections. Sterols 97-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 31273423-0 2019 Cytochrome P450 and Glutathione-S-Transferase Activity are Altered Following Environmentally Relevant Atrazine Exposures in Crayfish (Faxoniusvirilis). Atrazine 102-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-45 31763971-0 2021 Carbon-Carbon Bond Cleavage Catalyzed by Human Cytochrome P450 Enzymes: a-Ketol as the Key Intermediate Metabolite in Sequential Metabolism of Olanexidine. Carbon 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 31763971-0 2021 Carbon-Carbon Bond Cleavage Catalyzed by Human Cytochrome P450 Enzymes: a-Ketol as the Key Intermediate Metabolite in Sequential Metabolism of Olanexidine. olanexidine 143-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 31763971-5 2021 METHODS: A pair of regioisomers of alpha-ketol-containing C8 side chain olanexidine analogs (3,2-ketol olanexidine and 2,3-ketol olanexidine) were synthesized, followed by incubation in human liver microsomes, recombinant human cytochrome P450 enzymes or human hepatocytes, and subsequent metabolite identification using LC/UV/MS. fatty acid ketols 35-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 31763971-7 2021 Among 19 cytochrome P450 enzymes tested, CYP2D6, CYP3A4 and CYP3A5 were identified to catalyze carbon-carbon bond cleavage. Carbon 95-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-24 30557096-2 2019 Catalpol possesses numerous pharmacological activities, and however, little data available for the effects of catalpol on the activity of human liver cytochrome P450 (CYP) enzymes. catalpol 110-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-165 30557096-2 2019 Catalpol possesses numerous pharmacological activities, and however, little data available for the effects of catalpol on the activity of human liver cytochrome P450 (CYP) enzymes. catalpol 110-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-170 30557096-7 2019 The results indicated that catalpol could inhibit the activity of CYP3A4, CYP2E1 and CYP2C9, with IC50 values of 14.27, 22.4 and 14.69 muM, respectively, but those other CYP isoforms were not affected. catalpol 27-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 31418753-4 2019 The smoker"s paradox may be due to enhanced platelet response to clopidogrel therapy in active smokers as compared with nonsmokers caused by hepatic cytochrome P450 activation resulting in an increased generation of clopidogrel active metabolite. Clopidogrel 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-164 31418753-4 2019 The smoker"s paradox may be due to enhanced platelet response to clopidogrel therapy in active smokers as compared with nonsmokers caused by hepatic cytochrome P450 activation resulting in an increased generation of clopidogrel active metabolite. Clopidogrel 216-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-164 31515991-0 2019 In Vitro Metabolism of Auriculasin and Its Inhibitory Effects on Human Cytochrome P450 and UDP-Glucuronosyltransferase Enzymes. auriculasin 23-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 31515991-6 2019 In addition, CYP2D6, CYP2C9, CYP2C19, and CYP2C8 were the major CYP isoforms involved in the metabolism of auriculasin. auriculasin 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 31077555-1 2019 AIM: Imbalances in cytochrome P450 (CYP)-dependent eicosanoid formation may play a central role in ischemic acute kidney injury (AKI). Eicosanoids 51-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 31077555-1 2019 AIM: Imbalances in cytochrome P450 (CYP)-dependent eicosanoid formation may play a central role in ischemic acute kidney injury (AKI). Eicosanoids 51-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 31077555-5 2019 Circulating CYP-eicosanoid profiles were compared in patients who underwent cardiac surgery with (n = 21) and without (n = 38) developing postoperative AKI. Eicosanoids 16-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 31077555-12 2019 CONCLUSIONS: Pharmacological interventions targeting the CYP-eicosanoid pathway could offer promising new options for AKI prevention. Eicosanoids 61-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 31077555-13 2019 Individual differences in CYP-eicosanoid formation may contribute to the risk of developing AKI in clinical settings. Eicosanoids 30-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 31745121-11 2019 Among the enzymatic pathways that differentiate eicosanoid metabolites, we observed the highest prediction of overall preterm birth by lipoxygenase metabolites using random forest (AUC = 0.83 [0.69, 0.96]), followed by cytochrome p450 metabolites using adaptive elastic-net (AUC = 0.74 [0.52, 0.96]). Eicosanoids 48-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 219-234 31782268-1 2019 Fatty acid (FA)-derived lipid products generated by cytochrome P450 (CYP), lipoxygenase (LOX), and cyclo-oxygenase (COX) influence cardiovascular function. Fatty Acids 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 31782268-1 2019 Fatty acid (FA)-derived lipid products generated by cytochrome P450 (CYP), lipoxygenase (LOX), and cyclo-oxygenase (COX) influence cardiovascular function. Fatty Acids 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 31782268-8 2019 We used tandem mass spectrometry (LC-MS/MS) to measure epoxides derived from CYP monooxygenase, as well as metabolites derived from LOX, COX, and CYP hydroxylase pathways. Epoxy Compounds 55-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 31273423-3 2019 We examined sublethal effects of atrazine on the expression and activity of the detoxification enzymes cytochrome P450 (CYP450) and glutathione-S-transferase (GST) in crayfish. Atrazine 33-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 31273423-3 2019 We examined sublethal effects of atrazine on the expression and activity of the detoxification enzymes cytochrome P450 (CYP450) and glutathione-S-transferase (GST) in crayfish. Atrazine 33-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-126 31273423-6 2019 Atrazine exposure caused differential expression and activity of CYP450 and GST. Atrazine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-71 31375472-10 2019 Here, we report the application of MMHH in a high-throughput assay in a 384-well plate format for the evaluation of cytochrome P450 (P450) inhibition. mmhh 35-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 31430908-0 2019 In Vitro Inhibitory Effects of APINACA on Human Major Cytochrome P450, UDP-Glucuronosyltransferase Enzymes, and Drug Transporters. AKB48 31-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 31359235-5 2019 As the model drug, we chose diclofenac because of its known high CV toxicity, as markers of CV risks, we assessed concentrations of cytochrome P450-mediated metabolites of arachidonic acid (ArA), and we used adjuvant arthritis as an experimental model of arthritis. Arachidonic Acid 172-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-147 31469704-0 2019 Pharmacogenomics of Cytochrome P450 of Nimodipine Metabolism After Aneurysmal Subarachnoid Hemorrhage. Nimodipine 39-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 31469704-4 2019 METHODS AND RESULTS: This article reviews the CYP450 enzyme system including a review of the pharmacotherapy and pharmacogenomics of nimodipine for patients with aSAH illustrated with case study of a patient with abnormal drug metabolism. Nimodipine 133-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-52 29676195-0 2019 In vitro inhibitory effects of sophocarpine on human liver cytochrome P450 enzymes. sophocarpine 31-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 29676195-3 2019 However, whether sophocarpine affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. sophocarpine 17-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 29676195-3 2019 However, whether sophocarpine affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. sophocarpine 17-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 29676195-5 2019 In this study, the inhibitory effects of sophocarpine on the eight human liver CYP isoforms (CYP1A2, 3A4, 2A6, 2E1, 2D6, 2C9, 2C19, and 2C8) were investigated in vitro using human liver microsomes (HLMs). sophocarpine 41-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 29676195-7 2019 The results indicate that sophocarpine could inhibit the activity of CYP3A4 and 2C9, with the IC50 values of 12.22 and 15.96 muM, respectively, but that other CYP isoforms were not affected. sophocarpine 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 29676195-11 2019 The in vitro studies of sophocarpine with CYP isoforms suggested that sophocarpine has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP3A4 and 2C9. sophocarpine 70-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 31254976-1 2019 The spatial distributions of cytochrome P450 (CYP450) and glutathione (GSH) in liver lobules determine the heterogeneous hepatotoxicity of acetaminophen (APAP). Acetaminophen 139-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 31254976-1 2019 The spatial distributions of cytochrome P450 (CYP450) and glutathione (GSH) in liver lobules determine the heterogeneous hepatotoxicity of acetaminophen (APAP). Acetaminophen 139-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-52 31254976-1 2019 The spatial distributions of cytochrome P450 (CYP450) and glutathione (GSH) in liver lobules determine the heterogeneous hepatotoxicity of acetaminophen (APAP). Acetaminophen 154-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 31254976-1 2019 The spatial distributions of cytochrome P450 (CYP450) and glutathione (GSH) in liver lobules determine the heterogeneous hepatotoxicity of acetaminophen (APAP). Acetaminophen 154-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-52 31254976-5 2019 We found that the spatial distribution of GSH, glutathione S-transferases (GSTs), and CYP450 all contributes to the high acetaminophen protein adduct formation at zone 3 of the lobules. Acetaminophen 121-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-92 31254976-6 2019 The reversed spatial gradients of CYP450 and GSH cause quick depletion of GSH, which is further accelerated by the distribution of GST. Glutathione 74-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-40 31656654-3 2019 The cytochrome P450 (CYP) enzyme inducer, rifampicin, and the CYP inhibitor, clarithromycin, have clinical activity against MAC and key drugs in the treatment of MAC infection. Rifampin 42-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 31005596-3 2019 Tramadol labeling indicates cytochrome P450 (CYP) isozyme 2D6 ultrarapid metabolizer can produce dangerous (+)-M1 levels, and CYP2D6 poor metabolizers insufficient (+)-M1 for analgesia. Tramadol 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 31005596-3 2019 Tramadol labeling indicates cytochrome P450 (CYP) isozyme 2D6 ultrarapid metabolizer can produce dangerous (+)-M1 levels, and CYP2D6 poor metabolizers insufficient (+)-M1 for analgesia. Tramadol 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 31005596-7 2019 In trial B, CYP inhibition significantly depressed tramadol steady-state (+)-M1, reduced its adverse events, and led to insignificant analgesia comparable with placebo. Tramadol 51-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 31656654-3 2019 The cytochrome P450 (CYP) enzyme inducer, rifampicin, and the CYP inhibitor, clarithromycin, have clinical activity against MAC and key drugs in the treatment of MAC infection. Clarithromycin 77-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 30272491-0 2019 Development of new Coumarin-based profluorescent substrates for human cytochrome P450 enzymes. coumarin 19-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 30272491-4 2019 Molecular modeling indicated that 3-phenylcoumarin offers an excellent scaffold for the development of selective substrate compounds for various human CYP forms, as they could be metabolized to fluorescent 7-hydroxycoumarin derivatives. 3-Phenylcoumarin 34-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 30272491-4 2019 Molecular modeling indicated that 3-phenylcoumarin offers an excellent scaffold for the development of selective substrate compounds for various human CYP forms, as they could be metabolized to fluorescent 7-hydroxycoumarin derivatives. 7-hydroxycoumarin 206-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 30272491-5 2019 Oxidation of profluorescent coumarin derivatives to fluorescent metabolites by 13 important human liver xenobiotic-metabolizing CYP forms was determined by enzyme kinetic assays. coumarin 28-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 31296661-1 2019 Human cytochrome P450 (P450) 11B2 catalyzes the formation of aldosterone, the major endogenous human mineralocorticoid. Aldosterone 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-33 30973652-1 2019 Different views appear in the literature on the extent of specific cytochrome P450 (CYP) involvement in methadone metabolism. Methadone 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 31103741-5 2019 Our study aimed to analyze whether the PA representative lasiocarpine is toxified by human cytochrome P450 (CYP) enzymes. Pyrrolizidine Alkaloids 39-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 31103741-5 2019 Our study aimed to analyze whether the PA representative lasiocarpine is toxified by human cytochrome P450 (CYP) enzymes. Pyrrolizidine Alkaloids 39-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 31103741-5 2019 Our study aimed to analyze whether the PA representative lasiocarpine is toxified by human cytochrome P450 (CYP) enzymes. lasiocarpine 57-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 31103741-5 2019 Our study aimed to analyze whether the PA representative lasiocarpine is toxified by human cytochrome P450 (CYP) enzymes. lasiocarpine 57-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 31428108-6 2019 The enzymes that form the flavonoid scaffold structures probably first appeared by recruitment of enzymes from primary metabolic pathways, and later, enzymes that belong to superfamilies such as 2-oxoglutarate-dependent dioxygenase, cytochrome P450, and short-chain dehydrogenase/reductase modified and varied the structures. Flavonoids 26-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 233-289 30973652-1 2019 Different views appear in the literature on the extent of specific cytochrome P450 (CYP) involvement in methadone metabolism. Methadone 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 31212689-0 2019 In Vitro Human Metabolism and Inhibition Potency of Verbascoside for CYP Enzymes. acteoside 52-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 31333461-4 2019 Cytochrome P450 enzymes metabolize endogenous substrates (polyunsaturated fatty acids) to bioactive fatty acid epoxides that demonstrate biological activity with generally protective/anti-inflammatory and insulin-sensitizing effects. Fatty Acids, Unsaturated 58-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31333461-4 2019 Cytochrome P450 enzymes metabolize endogenous substrates (polyunsaturated fatty acids) to bioactive fatty acid epoxides that demonstrate biological activity with generally protective/anti-inflammatory and insulin-sensitizing effects. fatty acid epoxides 100-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 31111189-5 2019 Incubation of 21 recombinant human CYP enzymes with 0.5 microM IMQ and subsequent LC-MS analyses, in fact, identified CYP1A1 and CYP1A2 as being predominantly responsible for IMQ metabolism. Imiquimod 63-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 31029719-0 2019 Oxidative stress and cell death induction by amitraz and its metabolite BTS-27271 mediated through cytochrome P450 and NRF2 pathway alteration in primary hippocampal cell. Pyruvic Acid 72-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-114 31029719-3 2019 Amitraz was shown to induce different cytochrome P450 (CYP) isoenzymes involved with ROS and apoptotic cell death induction. amitraz 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 30964977-0 2019 Oxidation of Flavone, 5-Hydroxyflavone, and 5,7-Dihydroxyflavone to Mono-, Di-, and Tri-Hydroxyflavones by Human Cytochrome P450 Enzymes. 5-hydroxyflavone 22-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 30964977-0 2019 Oxidation of Flavone, 5-Hydroxyflavone, and 5,7-Dihydroxyflavone to Mono-, Di-, and Tri-Hydroxyflavones by Human Cytochrome P450 Enzymes. chrysin 44-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 30964977-0 2019 Oxidation of Flavone, 5-Hydroxyflavone, and 5,7-Dihydroxyflavone to Mono-, Di-, and Tri-Hydroxyflavones by Human Cytochrome P450 Enzymes. mono-, di-, and tri-hydroxyflavones 68-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 30964977-2 2019 In order to better understand the metabolism of these flavonoids in humans, we examined the oxidation of flavone, 5-hydroxyflavone (5OHF), and 57diOHF to various products by human cytochrome P450 (P450 or CYP) and liver microsomal enzymes. Flavonoids 54-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-195 30964977-2 2019 In order to better understand the metabolism of these flavonoids in humans, we examined the oxidation of flavone, 5-hydroxyflavone (5OHF), and 57diOHF to various products by human cytochrome P450 (P450 or CYP) and liver microsomal enzymes. flavone 105-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-195 30964977-2 2019 In order to better understand the metabolism of these flavonoids in humans, we examined the oxidation of flavone, 5-hydroxyflavone (5OHF), and 57diOHF to various products by human cytochrome P450 (P450 or CYP) and liver microsomal enzymes. 5-hydroxyflavone 114-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-195 30964977-2 2019 In order to better understand the metabolism of these flavonoids in humans, we examined the oxidation of flavone, 5-hydroxyflavone (5OHF), and 57diOHF to various products by human cytochrome P450 (P450 or CYP) and liver microsomal enzymes. 5ohf 132-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-195 30964977-2 2019 In order to better understand the metabolism of these flavonoids in humans, we examined the oxidation of flavone, 5-hydroxyflavone (5OHF), and 57diOHF to various products by human cytochrome P450 (P450 or CYP) and liver microsomal enzymes. 57diohf 143-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-195 31111189-3 2019 Treatment of human keratinocytes with 2.5 microM benzo[a]pyrene (BaP), a tobacco smoke constituent and aryl hydrocarbon receptor (AHR) agonist, for 24 h induced cytochrome P450 (CYP) 1A enzyme activity. Benzo(a)pyrene 49-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-176 31111189-3 2019 Treatment of human keratinocytes with 2.5 microM benzo[a]pyrene (BaP), a tobacco smoke constituent and aryl hydrocarbon receptor (AHR) agonist, for 24 h induced cytochrome P450 (CYP) 1A enzyme activity. Benzo(a)pyrene 49-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 178-181 31111189-3 2019 Treatment of human keratinocytes with 2.5 microM benzo[a]pyrene (BaP), a tobacco smoke constituent and aryl hydrocarbon receptor (AHR) agonist, for 24 h induced cytochrome P450 (CYP) 1A enzyme activity. Benzo(a)pyrene 65-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-176 31111189-3 2019 Treatment of human keratinocytes with 2.5 microM benzo[a]pyrene (BaP), a tobacco smoke constituent and aryl hydrocarbon receptor (AHR) agonist, for 24 h induced cytochrome P450 (CYP) 1A enzyme activity. Benzo(a)pyrene 65-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 178-181 31068367-0 2019 Interactions of Alectinib with Human ATP-Binding Cassette Drug Efflux Transporters and Cytochrome P450 Biotransformation Enzymes: Effect on Pharmacokinetic Multidrug Resistance. alectinib 16-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 31029719-3 2019 Amitraz was shown to induce different cytochrome P450 (CYP) isoenzymes involved with ROS and apoptotic cell death induction. amitraz 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 31029719-3 2019 Amitraz was shown to induce different cytochrome P450 (CYP) isoenzymes involved with ROS and apoptotic cell death induction. Reactive Oxygen Species 85-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 31029719-3 2019 Amitraz was shown to induce different cytochrome P450 (CYP) isoenzymes involved with ROS and apoptotic cell death induction. Reactive Oxygen Species 85-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 30964977-0 2019 Oxidation of Flavone, 5-Hydroxyflavone, and 5,7-Dihydroxyflavone to Mono-, Di-, and Tri-Hydroxyflavones by Human Cytochrome P450 Enzymes. flavone 13-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 31058221-3 2019 With oxidase catalysis as a model reaction, experimental studies and theoretical calculations revealed that single-atom nanozymes with carbon nanoframe-confined FeN5 active centers (FeN5 SA/CNF) catalytically behaved like the axial ligand-coordinated heme of cytochrome P450. Carbon 135-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 259-274 30981875-0 2019 Cytochrome P450 enzymes contribute to the metabolism of LSD to nor-LSD and 2-oxo-3-hydroxy-LSD: Implications for clinical LSD use. N-Demethyllysergic acid diethylamide 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 30981875-0 2019 Cytochrome P450 enzymes contribute to the metabolism of LSD to nor-LSD and 2-oxo-3-hydroxy-LSD: Implications for clinical LSD use. 2-oxo-3-hydroxylysergic acid diethylamide 75-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 30981875-3 2019 The aim of the current study was to identify the cytochrome P450 (CYP) isoforms involved in the metabolism of LSD to 6-norlysergic acid diethylamide (nor-LSD) and 2-oxo-3-hydroxy-LSD (O-H-LSD) in vitro, in order to evaluate potential effects of enzyme polymorphisms or prescription drugs on LSD pharmacokinetics. N-Demethyllysergic acid diethylamide 117-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 30981875-3 2019 The aim of the current study was to identify the cytochrome P450 (CYP) isoforms involved in the metabolism of LSD to 6-norlysergic acid diethylamide (nor-LSD) and 2-oxo-3-hydroxy-LSD (O-H-LSD) in vitro, in order to evaluate potential effects of enzyme polymorphisms or prescription drugs on LSD pharmacokinetics. N-Demethyllysergic acid diethylamide 117-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 30981875-3 2019 The aim of the current study was to identify the cytochrome P450 (CYP) isoforms involved in the metabolism of LSD to 6-norlysergic acid diethylamide (nor-LSD) and 2-oxo-3-hydroxy-LSD (O-H-LSD) in vitro, in order to evaluate potential effects of enzyme polymorphisms or prescription drugs on LSD pharmacokinetics. N-Demethyllysergic acid diethylamide 150-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 30981875-3 2019 The aim of the current study was to identify the cytochrome P450 (CYP) isoforms involved in the metabolism of LSD to 6-norlysergic acid diethylamide (nor-LSD) and 2-oxo-3-hydroxy-LSD (O-H-LSD) in vitro, in order to evaluate potential effects of enzyme polymorphisms or prescription drugs on LSD pharmacokinetics. N-Demethyllysergic acid diethylamide 150-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 30981875-3 2019 The aim of the current study was to identify the cytochrome P450 (CYP) isoforms involved in the metabolism of LSD to 6-norlysergic acid diethylamide (nor-LSD) and 2-oxo-3-hydroxy-LSD (O-H-LSD) in vitro, in order to evaluate potential effects of enzyme polymorphisms or prescription drugs on LSD pharmacokinetics. 2-oxo-3-hydroxylysergic acid diethylamide 163-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 30981875-3 2019 The aim of the current study was to identify the cytochrome P450 (CYP) isoforms involved in the metabolism of LSD to 6-norlysergic acid diethylamide (nor-LSD) and 2-oxo-3-hydroxy-LSD (O-H-LSD) in vitro, in order to evaluate potential effects of enzyme polymorphisms or prescription drugs on LSD pharmacokinetics. 2-oxo-3-hydroxylysergic acid diethylamide 163-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 30981875-3 2019 The aim of the current study was to identify the cytochrome P450 (CYP) isoforms involved in the metabolism of LSD to 6-norlysergic acid diethylamide (nor-LSD) and 2-oxo-3-hydroxy-LSD (O-H-LSD) in vitro, in order to evaluate potential effects of enzyme polymorphisms or prescription drugs on LSD pharmacokinetics. o-h-lsd 184-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 30981875-3 2019 The aim of the current study was to identify the cytochrome P450 (CYP) isoforms involved in the metabolism of LSD to 6-norlysergic acid diethylamide (nor-LSD) and 2-oxo-3-hydroxy-LSD (O-H-LSD) in vitro, in order to evaluate potential effects of enzyme polymorphisms or prescription drugs on LSD pharmacokinetics. o-h-lsd 184-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 30845890-2 2019 Amiodarone may also alter the pharmacokinetics of medications metabolized via cytochrome P450. Amiodarone 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 31091646-8 2019 SRIs are metabolized in the liver by several cytochrome P450 (CYP) enzymes. sris 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 31091646-8 2019 SRIs are metabolized in the liver by several cytochrome P450 (CYP) enzymes. sris 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 30918015-7 2019 Deactivation caused by DCA-induced cytochrome P450-cytochrome P420 conversion, as shown by the spectral titrations of isolated CYP3A proteins, was observed when DCA levels were near or higher than the critical micelle concentration (about 1500 muM). Deoxycholic Acid 23-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-66 30918015-7 2019 Deactivation caused by DCA-induced cytochrome P450-cytochrome P420 conversion, as shown by the spectral titrations of isolated CYP3A proteins, was observed when DCA levels were near or higher than the critical micelle concentration (about 1500 muM). Deoxycholic Acid 161-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-66 31151284-9 2019 AIs and N/DSs sharing the cytochrome P450 pathway are at risk of negative interactions. Nitrogen 8-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 31164617-3 2019 The metabolic rates of caffeine, dextromethorphan, midazolam, omeprazole, and Losartan to the CYP-specific metabolites are validated measures of in vivo CYP 1A2, 2D6, 3A4, 2C19, and 2C9 activities, respectively. Caffeine 23-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 31164617-3 2019 The metabolic rates of caffeine, dextromethorphan, midazolam, omeprazole, and Losartan to the CYP-specific metabolites are validated measures of in vivo CYP 1A2, 2D6, 3A4, 2C19, and 2C9 activities, respectively. Dextromethorphan 33-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 31164617-3 2019 The metabolic rates of caffeine, dextromethorphan, midazolam, omeprazole, and Losartan to the CYP-specific metabolites are validated measures of in vivo CYP 1A2, 2D6, 3A4, 2C19, and 2C9 activities, respectively. Midazolam 51-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 31164617-3 2019 The metabolic rates of caffeine, dextromethorphan, midazolam, omeprazole, and Losartan to the CYP-specific metabolites are validated measures of in vivo CYP 1A2, 2D6, 3A4, 2C19, and 2C9 activities, respectively. Omeprazole 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 31164617-3 2019 The metabolic rates of caffeine, dextromethorphan, midazolam, omeprazole, and Losartan to the CYP-specific metabolites are validated measures of in vivo CYP 1A2, 2D6, 3A4, 2C19, and 2C9 activities, respectively. Losartan 78-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 30888624-3 2019 The primary metabolic pathways for olanzapine are direct glucuronidation via uridine 5"-diphospho-glucuronosyltransferase (UGT)1A4 and cytochrome P450 (CYP)-mediated oxidation, mainly by CYP1A2. Olanzapine 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 31006007-10 2019 Cytochrome-P450-derived anti-inflammatory and cardioprotective epoxy-PUFAs increased linearly with n-3 PUFA dose and showed low interindividual variance (r2 > 0.95). epoxy-pufas 63-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 30888624-3 2019 The primary metabolic pathways for olanzapine are direct glucuronidation via uridine 5"-diphospho-glucuronosyltransferase (UGT)1A4 and cytochrome P450 (CYP)-mediated oxidation, mainly by CYP1A2. Olanzapine 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-155 30951643-1 2019 INTRODUCTION: Drugs used in HIV treatment; all protease inhibitors, some non-nucleoside reverse transcriptase inhibitors, and pharmacoenhancers ritonavir and cobicistat can inhibit cytochrome P450 (CYP) enzymes. Ritonavir 144-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-196 30951643-1 2019 INTRODUCTION: Drugs used in HIV treatment; all protease inhibitors, some non-nucleoside reverse transcriptase inhibitors, and pharmacoenhancers ritonavir and cobicistat can inhibit cytochrome P450 (CYP) enzymes. Ritonavir 144-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-201 30951643-1 2019 INTRODUCTION: Drugs used in HIV treatment; all protease inhibitors, some non-nucleoside reverse transcriptase inhibitors, and pharmacoenhancers ritonavir and cobicistat can inhibit cytochrome P450 (CYP) enzymes. Cobicistat 158-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-196 30951643-1 2019 INTRODUCTION: Drugs used in HIV treatment; all protease inhibitors, some non-nucleoside reverse transcriptase inhibitors, and pharmacoenhancers ritonavir and cobicistat can inhibit cytochrome P450 (CYP) enzymes. Cobicistat 158-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-201 30733060-0 2019 Cytochrome P450 enzymes: A driving force of plant diterpene diversity. Diterpenes 50-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 30733060-1 2019 In plant terpene biosynthesis, oxidation of the hydrocarbon backbone produced by terpene synthases is typically carried out by cytochrome P450 oxygenases (CYPs). Hydrocarbons 48-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-142 31278717-1 2019 Ketoconazole is a first orally available anti-fungal drug which has been reported as a potent inhibitor of human cytochrome P-450. Ketoconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 30733060-1 2019 In plant terpene biosynthesis, oxidation of the hydrocarbon backbone produced by terpene synthases is typically carried out by cytochrome P450 oxygenases (CYPs). Terpenes 9-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-142 30914192-0 2019 Ellipticine-loaded apoferritin nanocarrier retains DNA adduct-based cytochrome P450-facilitated toxicity in neuroblastoma cells. ellipticine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 30861250-3 2019 CYP2J2 is a membrane-bound cytochrome P450 in the heart involved in the metabolism of fatty acids and xenobiotics. Fatty Acids 86-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 30914192-6 2019 The interaction of ApoElli with microsomal membrane particles containing cytochrome P450 (CYP) biotransformation enzymes accelerated the release of ellipticine from this nanocarrier making it possible to be transferred into this membrane system even at pH 7.4 and facilitating CYP-mediated metabolism. ellipticine 148-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 30914192-6 2019 The interaction of ApoElli with microsomal membrane particles containing cytochrome P450 (CYP) biotransformation enzymes accelerated the release of ellipticine from this nanocarrier making it possible to be transferred into this membrane system even at pH 7.4 and facilitating CYP-mediated metabolism. ellipticine 148-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-93 30914192-6 2019 The interaction of ApoElli with microsomal membrane particles containing cytochrome P450 (CYP) biotransformation enzymes accelerated the release of ellipticine from this nanocarrier making it possible to be transferred into this membrane system even at pH 7.4 and facilitating CYP-mediated metabolism. ellipticine 148-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 277-280 30821969-5 2019 As an illustrative example with reliable data from previous GCMC simulations, absolute binding affinity of camphor to cytochrome P450 was calculated. Camphor 107-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 29779438-0 2019 Metabolism of deltamethrin and cis- and trans-permethrin by human expressed cytochrome P450 and carboxylesterase enzymes. decamethrin 14-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-112 29779438-0 2019 Metabolism of deltamethrin and cis- and trans-permethrin by human expressed cytochrome P450 and carboxylesterase enzymes. cis- and trans-permethrin 31-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-112 29779438-1 2019 The metabolism of the pyrethroids deltamethrin (DLM), cis-permethrin (CPM) and trans-permethrin (TPM) was studied in human expressed cytochrome P450 (CYP) and carboxylesterase (CES) enzymes. Pyrethrins 22-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 29779438-1 2019 The metabolism of the pyrethroids deltamethrin (DLM), cis-permethrin (CPM) and trans-permethrin (TPM) was studied in human expressed cytochrome P450 (CYP) and carboxylesterase (CES) enzymes. decamethrin 34-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 29779438-1 2019 The metabolism of the pyrethroids deltamethrin (DLM), cis-permethrin (CPM) and trans-permethrin (TPM) was studied in human expressed cytochrome P450 (CYP) and carboxylesterase (CES) enzymes. decamethrin 34-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 29779438-1 2019 The metabolism of the pyrethroids deltamethrin (DLM), cis-permethrin (CPM) and trans-permethrin (TPM) was studied in human expressed cytochrome P450 (CYP) and carboxylesterase (CES) enzymes. Permethrin 79-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 29779438-1 2019 The metabolism of the pyrethroids deltamethrin (DLM), cis-permethrin (CPM) and trans-permethrin (TPM) was studied in human expressed cytochrome P450 (CYP) and carboxylesterase (CES) enzymes. Permethrin 97-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 29779438-1 2019 The metabolism of the pyrethroids deltamethrin (DLM), cis-permethrin (CPM) and trans-permethrin (TPM) was studied in human expressed cytochrome P450 (CYP) and carboxylesterase (CES) enzymes. Permethrin 97-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 29779438-3 2019 Other CYP enzymes contributing to the metabolism of one or more of the three pyrethroids were CYP1A2, CYP2C8, CYP2C9*1, CYP2D6*1, CYP3A4 and CYP3A5. Pyrethrins 77-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 29779438-6 2019 Apparent CLint values for pyrethroid metabolism by CYP and CES enzymes were scaled to per gram of adult human liver using abundance values for microsomal CYP enzymes and for CES enzymes in liver microsomes and cytosol. Pyrethrins 26-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 29779438-8 2019 Due to their higher abundance, all three pyrethroids were extensively metabolised by CES enzymes in adult human liver, with CYP enzymes only accounting for 2%, 10% and 1% of total metabolism for DLM, CPM and TPM, respectively. Pyrethrins 41-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 29790809-5 2019 Cytochrome p450 (CYP) 2D6 was found to be the dominant CYP enzyme involved in the biotransformation of leelamine to its hydroxylated metabolite, whereas CYP2C19, CYP1A1, and CYP3A4 contributed to some extent. dehydroabietylamine 103-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29790809-5 2019 Cytochrome p450 (CYP) 2D6 was found to be the dominant CYP enzyme involved in the biotransformation of leelamine to its hydroxylated metabolite, whereas CYP2C19, CYP1A1, and CYP3A4 contributed to some extent. dehydroabietylamine 103-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 29790809-5 2019 Cytochrome p450 (CYP) 2D6 was found to be the dominant CYP enzyme involved in the biotransformation of leelamine to its hydroxylated metabolite, whereas CYP2C19, CYP1A1, and CYP3A4 contributed to some extent. dehydroabietylamine 103-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 30701961-1 2019 Human cytochrome P450 (P450) family 4 enzymes are involved in the metabolism of fatty acids and the bioactivation of carcinogenic arylamines and toxic natural products, e.g., 4-ipomeanol. Fatty Acids 80-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-37 30936195-5 2019 Although variants were reported for all drugs, the highest level of evidence was found in cytochrome P450 (CYP450) genotype variation related to clopidogrel response. Clopidogrel 145-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 30936195-5 2019 Although variants were reported for all drugs, the highest level of evidence was found in cytochrome P450 (CYP450) genotype variation related to clopidogrel response. Clopidogrel 145-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-113 30998396-1 2019 Common genetic variation in CYP2C19 (cytochrome P450, family 2, subfamily C, polypeptide 19) *2 and *3 alleles leads to a loss of functional protein, and carriers of these loss-of-function alleles when treated with clopidogrel have significantly reduced clopidogrel active metabolite levels and high on-treatment platelet reactivity resulting in increased risk of major adverse cardiovascular events, especially after percutaneous coronary intervention. Clopidogrel 215-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-62 30998396-1 2019 Common genetic variation in CYP2C19 (cytochrome P450, family 2, subfamily C, polypeptide 19) *2 and *3 alleles leads to a loss of functional protein, and carriers of these loss-of-function alleles when treated with clopidogrel have significantly reduced clopidogrel active metabolite levels and high on-treatment platelet reactivity resulting in increased risk of major adverse cardiovascular events, especially after percutaneous coronary intervention. Clopidogrel 254-265 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-62 30701961-1 2019 Human cytochrome P450 (P450) family 4 enzymes are involved in the metabolism of fatty acids and the bioactivation of carcinogenic arylamines and toxic natural products, e.g., 4-ipomeanol. arylamines 130-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-37 30701961-1 2019 Human cytochrome P450 (P450) family 4 enzymes are involved in the metabolism of fatty acids and the bioactivation of carcinogenic arylamines and toxic natural products, e.g., 4-ipomeanol. 4-ipomeanol 175-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-37 29987837-9 2019 Cytochrome P450-mediated biotransformation of cladribine is of minor importance. Cladribine 46-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 30428505-2 2019 Oprozomib is a rare example of a small molecule drug that demonstrates cytochrome P450 (CYP) mRNA suppression. ONX 0912 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 30428505-2 2019 Oprozomib is a rare example of a small molecule drug that demonstrates cytochrome P450 (CYP) mRNA suppression. ONX 0912 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 30689138-4 2019 Since the induction of certain cytochrome P450 (CYP) enzymes accelerates warfarin metabolism, using CYP inducers, such as phenobarbital, to accelerate brodifacoum clearance seems plausible. Warfarin 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 30689138-4 2019 Since the induction of certain cytochrome P450 (CYP) enzymes accelerates warfarin metabolism, using CYP inducers, such as phenobarbital, to accelerate brodifacoum clearance seems plausible. Warfarin 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 30689138-4 2019 Since the induction of certain cytochrome P450 (CYP) enzymes accelerates warfarin metabolism, using CYP inducers, such as phenobarbital, to accelerate brodifacoum clearance seems plausible. Warfarin 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 30689138-4 2019 Since the induction of certain cytochrome P450 (CYP) enzymes accelerates warfarin metabolism, using CYP inducers, such as phenobarbital, to accelerate brodifacoum clearance seems plausible. Phenobarbital 122-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 30689138-4 2019 Since the induction of certain cytochrome P450 (CYP) enzymes accelerates warfarin metabolism, using CYP inducers, such as phenobarbital, to accelerate brodifacoum clearance seems plausible. Phenobarbital 122-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 30689138-4 2019 Since the induction of certain cytochrome P450 (CYP) enzymes accelerates warfarin metabolism, using CYP inducers, such as phenobarbital, to accelerate brodifacoum clearance seems plausible. Phenobarbital 122-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 30976185-3 2019 The effect of pterostilbene on cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzyme activities were studied using the enzyme-selective substrates amodiaquine (CYP2C8), midazolam (CYP3A4), estradiol (UGT1A1), serotonin (UGT1A6) and mycophenolic acid (UGT1A8/9/10). pterostilbene 14-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 30467864-0 2019 Effect of an anxiolytic botanical containing Souroubea sympetala and Platanus occidentalis on in-vitro diazepam human cytochrome P450-mediated metabolism. Diazepam 103-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 30976185-3 2019 The effect of pterostilbene on cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzyme activities were studied using the enzyme-selective substrates amodiaquine (CYP2C8), midazolam (CYP3A4), estradiol (UGT1A1), serotonin (UGT1A6) and mycophenolic acid (UGT1A8/9/10). pterostilbene 14-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 30976185-3 2019 The effect of pterostilbene on cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzyme activities were studied using the enzyme-selective substrates amodiaquine (CYP2C8), midazolam (CYP3A4), estradiol (UGT1A1), serotonin (UGT1A6) and mycophenolic acid (UGT1A8/9/10). Amodiaquine 160-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 30976185-3 2019 The effect of pterostilbene on cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzyme activities were studied using the enzyme-selective substrates amodiaquine (CYP2C8), midazolam (CYP3A4), estradiol (UGT1A1), serotonin (UGT1A6) and mycophenolic acid (UGT1A8/9/10). Amodiaquine 160-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 30389730-0 2019 Characterization of the In Vitro Inhibitory Potential of the Oligonucleotide Imetelstat on Human Cytochrome P450 Enzymes with Predictions of In Vivo Drug-Drug Interactions. Oligonucleotides 61-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-112 30860683-13 2019 CONCLUSION: The results found demonstrate that TKIs vandetanib, lenvatinib and cabozantinib cause a decrease in oxidative activation of DNA-damaging drug ellipticine by several CYP enzymes in vitro which might lead to a decrease in its pharmacological efficiency. ellipticine 154-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 30247644-10 2019 This was supported by significantly increased transcriptional levels of 5-lipoxygenase and CYP4F3 in whole blood cells from AG patients compared with those of controls, and the uric acid-caused dose-relevant and time-dependent activation of 5-lipoxygenase and CYP4F3 at the transcriptional and molecular levels in vitro. Uric Acid 177-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-97 30247644-10 2019 This was supported by significantly increased transcriptional levels of 5-lipoxygenase and CYP4F3 in whole blood cells from AG patients compared with those of controls, and the uric acid-caused dose-relevant and time-dependent activation of 5-lipoxygenase and CYP4F3 at the transcriptional and molecular levels in vitro. Uric Acid 177-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 260-266 30633519-2 2019 CYP450 is a nanomachine that uses dioxygen and two reducing and two proton equivalents to oxidize a plethora of molecules (so-called substrates) as a means of supplying bio-organisms with essential molecules (e.g., brain neurotransmitters, sex hormones, etc.) nanomachine 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 30633519-2 2019 CYP450 is a nanomachine that uses dioxygen and two reducing and two proton equivalents to oxidize a plethora of molecules (so-called substrates) as a means of supplying bio-organisms with essential molecules (e.g., brain neurotransmitters, sex hormones, etc.) Oxygen 34-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 30633519-7 2019 CYP450 is a heme enzyme that contains a ferric ion (FeIII) coordinated by a porphyrin ligand, a water molecule, and a cysteinate ligand that is provided by a strategic residue of the encapsulating protein. Heme 12-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 30633519-7 2019 CYP450 is a heme enzyme that contains a ferric ion (FeIII) coordinated by a porphyrin ligand, a water molecule, and a cysteinate ligand that is provided by a strategic residue of the encapsulating protein. Porphyrins 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 30633519-7 2019 CYP450 is a heme enzyme that contains a ferric ion (FeIII) coordinated by a porphyrin ligand, a water molecule, and a cysteinate ligand that is provided by a strategic residue of the encapsulating protein. Water 96-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 30633519-7 2019 CYP450 is a heme enzyme that contains a ferric ion (FeIII) coordinated by a porphyrin ligand, a water molecule, and a cysteinate ligand that is provided by a strategic residue of the encapsulating protein. cysteinate 118-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 30399481-1 2019 Cytochrome P450 (CYP) is a class of heme-containing enzymes which mainly catalyze a monooxygenation reaction of various chemicals, and hence CYP plays a key role in the drug metabolism. Heme 36-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 30399481-1 2019 Cytochrome P450 (CYP) is a class of heme-containing enzymes which mainly catalyze a monooxygenation reaction of various chemicals, and hence CYP plays a key role in the drug metabolism. Heme 36-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 30399481-1 2019 Cytochrome P450 (CYP) is a class of heme-containing enzymes which mainly catalyze a monooxygenation reaction of various chemicals, and hence CYP plays a key role in the drug metabolism. Heme 36-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-144 30399481-2 2019 Although CYP2C19 isoform is a minor hepatic CYP, it metabolizes clinically important drugs such as omeprazole and S-mephenytoin. Omeprazole 99-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 30399481-2 2019 Although CYP2C19 isoform is a minor hepatic CYP, it metabolizes clinically important drugs such as omeprazole and S-mephenytoin. Mephenytoin 114-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 30860683-7 2019 METHODS: Ellipticine oxidation catalyzed by hepatic microsomes, recombinant CYP enzymes and peroxidases (horseradish peroxidase, lactoperoxidase and myeloperoxidase) and the effect of TKIs (vandetanib, lenvatinib and cabozantinib) on this oxidation were analyzed by HPLC used for separation of ellipticine metabolites and quantification of their amounts formed during oxidation. ellipticine 9-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 30860683-8 2019 RESULTS: The CYP enzymatic system oxidizes ellipticine up to five metabolites (9-hydroxy-, 12-hydroxy-, 13-hydroxy-, 7-hydroxyellipticine, and ellipticine N2- oxide), while peroxidases form predominantly ellipticine dimer. ellipticine 43-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 30860683-8 2019 RESULTS: The CYP enzymatic system oxidizes ellipticine up to five metabolites (9-hydroxy-, 12-hydroxy-, 13-hydroxy-, 7-hydroxyellipticine, and ellipticine N2- oxide), while peroxidases form predominantly ellipticine dimer. 9-hydroxy-, 12-hydroxy-, 13-hydroxy-, 7-hydroxyellipticine 79-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 30860683-8 2019 RESULTS: The CYP enzymatic system oxidizes ellipticine up to five metabolites (9-hydroxy-, 12-hydroxy-, 13-hydroxy-, 7-hydroxyellipticine, and ellipticine N2- oxide), while peroxidases form predominantly ellipticine dimer. ellipticine n2- oxide 143-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 30860683-8 2019 RESULTS: The CYP enzymatic system oxidizes ellipticine up to five metabolites (9-hydroxy-, 12-hydroxy-, 13-hydroxy-, 7-hydroxyellipticine, and ellipticine N2- oxide), while peroxidases form predominantly ellipticine dimer. ellipticine 126-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 30860683-10 2019 Of individual CYP enzymes catalyzing oxidation of ellipticine, TKIs inhibited oxidation of ellipticine catalyzed by CYP2D6 > 2D1 > 2C9 > 3A1 > 3A4, the CYP enzymes participating in ellipticine oxidation to metabolites increasing the ellipticine anticancer efficiency. ellipticine 50-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 30860683-10 2019 Of individual CYP enzymes catalyzing oxidation of ellipticine, TKIs inhibited oxidation of ellipticine catalyzed by CYP2D6 > 2D1 > 2C9 > 3A1 > 3A4, the CYP enzymes participating in ellipticine oxidation to metabolites increasing the ellipticine anticancer efficiency. ellipticine 50-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 30860683-10 2019 Of individual CYP enzymes catalyzing oxidation of ellipticine, TKIs inhibited oxidation of ellipticine catalyzed by CYP2D6 > 2D1 > 2C9 > 3A1 > 3A4, the CYP enzymes participating in ellipticine oxidation to metabolites increasing the ellipticine anticancer efficiency. ellipticine 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 30860683-10 2019 Of individual CYP enzymes catalyzing oxidation of ellipticine, TKIs inhibited oxidation of ellipticine catalyzed by CYP2D6 > 2D1 > 2C9 > 3A1 > 3A4, the CYP enzymes participating in ellipticine oxidation to metabolites increasing the ellipticine anticancer efficiency. ellipticine 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 30860683-10 2019 Of individual CYP enzymes catalyzing oxidation of ellipticine, TKIs inhibited oxidation of ellipticine catalyzed by CYP2D6 > 2D1 > 2C9 > 3A1 > 3A4, the CYP enzymes participating in ellipticine oxidation to metabolites increasing the ellipticine anticancer efficiency. ellipticine 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 30860683-10 2019 Of individual CYP enzymes catalyzing oxidation of ellipticine, TKIs inhibited oxidation of ellipticine catalyzed by CYP2D6 > 2D1 > 2C9 > 3A1 > 3A4, the CYP enzymes participating in ellipticine oxidation to metabolites increasing the ellipticine anticancer efficiency. ellipticine 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 30860683-10 2019 Of individual CYP enzymes catalyzing oxidation of ellipticine, TKIs inhibited oxidation of ellipticine catalyzed by CYP2D6 > 2D1 > 2C9 > 3A1 > 3A4, the CYP enzymes participating in ellipticine oxidation to metabolites increasing the ellipticine anticancer efficiency. ellipticine 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 30860683-10 2019 Of individual CYP enzymes catalyzing oxidation of ellipticine, TKIs inhibited oxidation of ellipticine catalyzed by CYP2D6 > 2D1 > 2C9 > 3A1 > 3A4, the CYP enzymes participating in ellipticine oxidation to metabolites increasing the ellipticine anticancer efficiency. ellipticine 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 30860683-13 2019 CONCLUSION: The results found demonstrate that TKIs vandetanib, lenvatinib and cabozantinib cause a decrease in oxidative activation of DNA-damaging drug ellipticine by several CYP enzymes in vitro which might lead to a decrease in its pharmacological efficiency. vandetanib 52-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 30860683-13 2019 CONCLUSION: The results found demonstrate that TKIs vandetanib, lenvatinib and cabozantinib cause a decrease in oxidative activation of DNA-damaging drug ellipticine by several CYP enzymes in vitro which might lead to a decrease in its pharmacological efficiency. lenvatinib 64-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 30860683-13 2019 CONCLUSION: The results found demonstrate that TKIs vandetanib, lenvatinib and cabozantinib cause a decrease in oxidative activation of DNA-damaging drug ellipticine by several CYP enzymes in vitro which might lead to a decrease in its pharmacological efficiency. cabozantinib 79-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 31576541-6 2019 We also review the updated roles of the ethanol-inducible cytochrome P450-2E1 (CYP2E1) and other cytochrome P450 isozymes in the metabolism of various potentially toxic substrates, and consequent toxicities, including carcinogenesis in different tissues. Ethanol 40-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 30828066-0 2019 Prediction of Human Hepatic Clearance for Cytochrome P450 Substrates via a New Culture Method Using the Collagen Vitrigel Membrane Chamber and Fresh Hepatocytes Isolated from Liver Humanized Mice. vitrigel 113-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 30499388-0 2019 Biology of Heme: Drug Interactions and Adverse Drug Reactions with CYP450. Heme 11-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-73 30389730-3 2019 Other oligonucleotides have been previously shown to have in vitro test-system-dependent outcomes when potent cytochrome P450 inhibition in human liver microsomes (HLM) is observed, but such inhibition is not observed in cryopreserved human hepatocytes (CHH). Oligonucleotides 6-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 30788785-6 2019 We also describe the measurement of the activity of the cytochrome P450 expressed by taking the example of cytochrome P450 2J2, the primary P450 expressed in the human heart and CYP725A4, the primary cytochrome P450 expressed in the first step of taxol synthesis. Paclitaxel 247-252 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 30371861-0 2019 Identification of Cytochrome P450 Polymorphisms in Burn Patients and Impact on Fentanyl Pharmacokinetics: A Pilot Study. Fentanyl 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 30371861-6 2019 Functional CYP polymorphisms can significantly alter opiate levels resulting in inadequate analgesia or life-threatening toxicity. Opiate Alkaloids 53-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-14 30151903-1 2019 Acetaminophen (APAP)-induced liver injury is initiated by metabolism of APAP by the cytochrome P-450 (CYP) system, primarily CYP2E1. Acetaminophen 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 30151903-1 2019 Acetaminophen (APAP)-induced liver injury is initiated by metabolism of APAP by the cytochrome P-450 (CYP) system, primarily CYP2E1. Acetaminophen 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 30151903-1 2019 Acetaminophen (APAP)-induced liver injury is initiated by metabolism of APAP by the cytochrome P-450 (CYP) system, primarily CYP2E1. Acetaminophen 15-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 30151903-1 2019 Acetaminophen (APAP)-induced liver injury is initiated by metabolism of APAP by the cytochrome P-450 (CYP) system, primarily CYP2E1. Acetaminophen 15-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 30151903-2 2019 We previously demonstrated CYP inhibition following administration of a liquid APAP formulation containing propylene glycol, a CYP2E1 inhibitor, and other excipients. Acetaminophen 79-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 30151903-2 2019 We previously demonstrated CYP inhibition following administration of a liquid APAP formulation containing propylene glycol, a CYP2E1 inhibitor, and other excipients. Propylene Glycol 107-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 30151903-3 2019 This study was undertaken to determine if propylene glycol specifically inhibits production of CYP-derived metabolites and if propylene glycol reduces the rise in alanine aminotransferase (ALT) seen following prolonged APAP dosing. Propylene Glycol 42-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-98 30151903-8 2019 For all subjects, the mean percentage of CYP-derived metabolites produced was 5.8% (APAP) versus 4.3% (APAP + propylene glycol); P = .018. Acetaminophen 84-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 30151903-8 2019 For all subjects, the mean percentage of CYP-derived metabolites produced was 5.8% (APAP) versus 4.3% (APAP + propylene glycol); P = .018. Acetaminophen 103-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 30151903-8 2019 For all subjects, the mean percentage of CYP-derived metabolites produced was 5.8% (APAP) versus 4.3% (APAP + propylene glycol); P = .018. Propylene Glycol 110-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 30151903-9 2019 This effect was solely attributable to the responders: the mean percentage of CYP metabolites of responders was 7.7% (APAP) versus 4.6% (APAP + propylene glycol), P = .050, whereas there was no difference for the nonresponders. Acetaminophen 118-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 30788785-6 2019 We also describe the measurement of the activity of the cytochrome P450 expressed by taking the example of cytochrome P450 2J2, the primary P450 expressed in the human heart and CYP725A4, the primary cytochrome P450 expressed in the first step of taxol synthesis. Paclitaxel 247-252 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 30544127-0 2019 In vitro Inhibitory Effects of Isofraxidin on Human Liver Cytochrome P450 Enzymes. isofraxidin 31-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 30544127-2 2019 However, whether isofraxidin affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. isofraxidin 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 30544127-7 2019 The in vitro studies of isofraxidin with CYP isoforms indicate that isofraxidin has the potential to cause pharmacokinetic drug interactions with other coadministered drugs metabolized by -CYP1A2, 3A4, and 2E1. isofraxidin 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 30544127-2 2019 However, whether isofraxidin affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. isofraxidin 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 30544127-7 2019 The in vitro studies of isofraxidin with CYP isoforms indicate that isofraxidin has the potential to cause pharmacokinetic drug interactions with other coadministered drugs metabolized by -CYP1A2, 3A4, and 2E1. isofraxidin 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 31587003-8 2019 CONCLUSION: The in vitro studies of cynaroside with CYP isoforms indicate that cynaroside has the potential to cause pharmacokinetic drug interactions with other coadministered drugs metabolized by CYP1A2, 3A4, and 2C9. luteolin-7-glucoside 36-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 31587003-2 2019 However, whether cynaroside affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. luteolin-7-glucoside 17-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 31587003-2 2019 However, whether cynaroside affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. luteolin-7-glucoside 17-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 30544127-3 2019 In this study, the inhibitory effects of isofraxidin on the 8 human liver CYP isoforms (i.e., 1A2, 3A4, 2A6, 2E1, 2D6, 2C9, 2C19, and 2C8) were investigated in vitro using human liver microsomes. isofraxidin 41-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 31587003-3 2019 The purpose of this study was to investigate the effects of cynaroside on 8 major CYP isoforms in human liver microsomes (HLMs). luteolin-7-glucoside 60-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 31587003-8 2019 CONCLUSION: The in vitro studies of cynaroside with CYP isoforms indicate that cynaroside has the potential to cause pharmacokinetic drug interactions with other coadministered drugs metabolized by CYP1A2, 3A4, and 2C9. luteolin-7-glucoside 79-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 30544127-4 2019 The results showed that isofraxidin inhibited the activity of CYP1A2, 3A4, and 2E1, with IC50 values of 23.01, 15.49, and 15.98 micromol/L, respectively, but that other CYP isoforms were not affected. isofraxidin 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 31587003-4 2019 METHODS: In this study, the inhibitory effects of cynaroside on the 8 human liver CYP isoforms (i.e., 1A2, 3A4, 2A6, 2E1, 2D6, 2C9, 2C19, and 2C8) were investigated in vitro using HLMs. luteolin-7-glucoside 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 31587003-5 2019 RESULTS: The results showed that cynaroside inhibited the activity of CYP1A2, 3A4, and 2C9, with IC50 values of 21.74, 15.88, and 16.58 mumol/L, respectively, but that other CYP isoforms were not affected. luteolin-7-glucoside 33-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 30576387-5 2018 Activation of the AHR leads to upregulation of cytochrome P450 (CYP) metabolizing enzymes which are important for the biotransformation of toxicants to less toxic, or in the case of PAHs, more toxic intermediates. Polycyclic Aromatic Hydrocarbons 182-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 30560267-0 2018 Role of cytochrome P450 epoxygenase-dependent arachidonic acid metabolites in kidney physiology and diseases. Arachidonic Acid 46-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 30560267-3 2018 Pharmacological and biochemical tools have been used to conduct detailed studies on the metabolization of arachidonic acid by cytochrome P450 (CYP450) in renal microvasculature. Arachidonic Acid 106-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-141 30560267-3 2018 Pharmacological and biochemical tools have been used to conduct detailed studies on the metabolization of arachidonic acid by cytochrome P450 (CYP450) in renal microvasculature. Arachidonic Acid 106-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-149 30560267-4 2018 CYP450 epoxygenase metabolites epoxyeicosatrienoic acids (EETs) are mainly produced in renal microvessels. epoxyeicosatrienoic acids 31-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 30560267-4 2018 CYP450 epoxygenase metabolites epoxyeicosatrienoic acids (EETs) are mainly produced in renal microvessels. eets 58-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 30583596-5 2018 In general, cannabinoids are usually well tolerated, but bidirectional effects may be expected with concomitant administered agents via affected membrane transporters (Glycoprotein p, breast cancer resistance proteins, and multidrug resistance proteins) and metabolizing enzymes (Cytochrome P450 and UDP-glucuronosyltransferases). Cannabinoids 12-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 280-295 30576387-5 2018 Activation of the AHR leads to upregulation of cytochrome P450 (CYP) metabolizing enzymes which are important for the biotransformation of toxicants to less toxic, or in the case of PAHs, more toxic intermediates. Polycyclic Aromatic Hydrocarbons 182-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 30576387-11 2018 SRM1649b PAH mixtures enhanced Th17 differentiation in an AHR-dependent but CYP-independent manner and increased the percent of IFNgamma positive DCs. Polycyclic Aromatic Hydrocarbons 9-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 30422622-2 2018 Herein, a novel light-driven enzymatic bionanoreactor is designed with the gold nanoparticle (NP)-modified carbon nanocage (Au@CNC) as a nanoreactor and meso-tetrakis(4-carboxyphenyl)porphyrin (TCPP) as a photosensitizer for cytochrome P450-mediated drug metabolism. meso 153-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 225-240 30587978-10 2018 Conclusion: Our findings revealed that DN, GO and GN might interfere with xenobiotic and drug metabolism in the liver by interactions with CYP isoenzymes responsible for the process. dn 39-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 30587978-10 2018 Conclusion: Our findings revealed that DN, GO and GN might interfere with xenobiotic and drug metabolism in the liver by interactions with CYP isoenzymes responsible for the process. graphene oxide 43-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 30205091-2 2018 Methadone undergoes N-demethylation by multiple cytochrome P450 (CYP) enzymes including CYP3A4, CYP2B6, CYP2C19, CYP2D6, CYP2C9, and CYP2C8. Methadone 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 30587978-10 2018 Conclusion: Our findings revealed that DN, GO and GN might interfere with xenobiotic and drug metabolism in the liver by interactions with CYP isoenzymes responsible for the process. gossypolone 50-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 30422622-0 2018 Enhanced Metabolic Activity of Cytochrome P450 via Carbon Nanocage-Based Photochemical Bionanoreactor. Carbon 51-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 30422622-2 2018 Herein, a novel light-driven enzymatic bionanoreactor is designed with the gold nanoparticle (NP)-modified carbon nanocage (Au@CNC) as a nanoreactor and meso-tetrakis(4-carboxyphenyl)porphyrin (TCPP) as a photosensitizer for cytochrome P450-mediated drug metabolism. Gold 124-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 225-240 29858835-4 2018 Most drug interaction reports with CAM focus on cytochrome P450 (CYP) modulation. cafestol palmitate 35-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 29858835-4 2018 Most drug interaction reports with CAM focus on cytochrome P450 (CYP) modulation. cafestol palmitate 35-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 30205091-2 2018 Methadone undergoes N-demethylation by multiple cytochrome P450 (CYP) enzymes including CYP3A4, CYP2B6, CYP2C19, CYP2D6, CYP2C9, and CYP2C8. Methadone 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 30205091-2 2018 Methadone undergoes N-demethylation by multiple cytochrome P450 (CYP) enzymes including CYP3A4, CYP2B6, CYP2C19, CYP2D6, CYP2C9, and CYP2C8. Nitrogen 20-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 30205091-2 2018 Methadone undergoes N-demethylation by multiple cytochrome P450 (CYP) enzymes including CYP3A4, CYP2B6, CYP2C19, CYP2D6, CYP2C9, and CYP2C8. Nitrogen 20-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 30205091-5 2018 In general, CYP inhibitors altered methadone exposure with no adverse effects. Methadone 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 30205091-6 2018 CYP inducers generally decreased methadone exposure with some reports of withdrawal symptoms in the subjects. Methadone 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 30205091-8 2018 For certain antiviral medicines which are dual inhibitor(s) and inducer(s) for CYP enzymes, their effect on methadone pharmacokinetics can change with time since the effect of induction is usually delayed compared to the effect of inhibition. Methadone 108-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 29935100-1 2018 BACKGROUND & AIMS: Fatty liver disease is an important complication associated with liver transplantation, and the cytochrome P-450 system of the donor liver may be involved in its pathogenesis. Adenosine Monophosphate 12-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 31070542-9 2018 DISCUSSION AND CONCLUSIONS: The in vitro studies of bergenin with CYP isoforms indicate that bergenin has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP3A4, 2E1 and 2C9. bergenin 93-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 30280198-1 2018 Cytochrome P450 family 4 (CYP4) enzymes are known as microsomal omega (omega)-hydroxylases that metabolize fatty acids, eicosanoids, vitamin D and carcinogens. Fatty Acids 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-24 30280198-1 2018 Cytochrome P450 family 4 (CYP4) enzymes are known as microsomal omega (omega)-hydroxylases that metabolize fatty acids, eicosanoids, vitamin D and carcinogens. Fatty Acids 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-30 30280198-1 2018 Cytochrome P450 family 4 (CYP4) enzymes are known as microsomal omega (omega)-hydroxylases that metabolize fatty acids, eicosanoids, vitamin D and carcinogens. Eicosanoids 120-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-24 30280198-1 2018 Cytochrome P450 family 4 (CYP4) enzymes are known as microsomal omega (omega)-hydroxylases that metabolize fatty acids, eicosanoids, vitamin D and carcinogens. Eicosanoids 120-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-30 30280198-1 2018 Cytochrome P450 family 4 (CYP4) enzymes are known as microsomal omega (omega)-hydroxylases that metabolize fatty acids, eicosanoids, vitamin D and carcinogens. Vitamin D 133-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-24 30280198-1 2018 Cytochrome P450 family 4 (CYP4) enzymes are known as microsomal omega (omega)-hydroxylases that metabolize fatty acids, eicosanoids, vitamin D and carcinogens. Vitamin D 133-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-30 29964123-4 2018 Small molecule compounds targeting enzymes and CYP epoxy-fatty acid metabolites have evolved rapidly over the last two decades. Fatty Acids 57-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 30048707-1 2018 20-HETE, the omega-hydroxylation product of arachidonic acid catalyzed by enzymes of the cytochrome P450 (CYP) 4A and 4F gene families, is a bioactive lipid mediator with potent effects on the vasculature including stimulation of smooth muscle cell contractility, migration and proliferation as well as activation of endothelial cell dysfunction and inflammation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-120 30607725-0 2018 A New Method for Quantitative Determination of Steroid Metabolites of Cytochrome P450-Dependent Reactions Using Fluorescent Spectroscopy. Steroids 47-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 30048707-1 2018 20-HETE, the omega-hydroxylation product of arachidonic acid catalyzed by enzymes of the cytochrome P450 (CYP) 4A and 4F gene families, is a bioactive lipid mediator with potent effects on the vasculature including stimulation of smooth muscle cell contractility, migration and proliferation as well as activation of endothelial cell dysfunction and inflammation. Arachidonic Acid 44-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-120 28385095-0 2018 In vitro inhibitory effects of pristimerin on human liver cytochrome P450 enzymes. pristimerin 31-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 30285425-6 2018 On a separate note, the cytochrome P450 (CYP) epoxygenase CYP2J2 is the most abundant CYP in the heart where it catalyzes the metabolism of AA and AA-derived eCBs to bioactive epoxides that are involved in diverse cardiovascular functions. ecbs 158-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-63 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. epoxyeicosatrienoic acids 76-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. epoxyeicosatrienoic acids 76-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-63 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. eets 103-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. eets 103-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-63 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 113-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 113-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-63 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 146-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 30400671-2 2018 Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP450) enzymes to epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), vasoactive and natriuretic metabolites that contribute to blood pressure (BP) regulation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 146-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-63 30400671-3 2018 Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) omega-3 polyunsaturated fatty acids may compete with AA for CYP450-dependent bioactive lipid mediator formation. Eicosapentaenoic Acid 0-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-125 30400671-3 2018 Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) omega-3 polyunsaturated fatty acids may compete with AA for CYP450-dependent bioactive lipid mediator formation. Eicosapentaenoic Acid 23-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-125 30400671-3 2018 Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) omega-3 polyunsaturated fatty acids may compete with AA for CYP450-dependent bioactive lipid mediator formation. Docosahexaenoic Acids 32-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-125 30400671-3 2018 Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) omega-3 polyunsaturated fatty acids may compete with AA for CYP450-dependent bioactive lipid mediator formation. Docosahexaenoic Acids 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-125 30400671-3 2018 Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) omega-3 polyunsaturated fatty acids may compete with AA for CYP450-dependent bioactive lipid mediator formation. omega-3 polyunsaturated fatty acids 59-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-125 30400671-5 2018 Fatty acid profile moderately correlated with the corresponding CYP450-derived metabolites but their levels did not differ between children with normal BP (NBP) and high BP (HBP), except for higher EPA-derived epoxyeicosatetraenoic acids (EEQs) and their diols in HBP group, in which also the estimated CYP450-epoxygenase activity was higher. Fatty Acids 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-70 30259074-3 2018 Therefore, this study was designed to develop CYP4F selective inhibitors using a novel inhibitory assay of 20-HETE formation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 107-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-51 30122094-0 2018 In vitro inhibitory effects of Friedelin on human liver cytochrome P450 enzymes. friedelin 31-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 30122094-2 2018 However, the affects of Friedelin on the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. friedelin 24-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 30122094-2 2018 However, the affects of Friedelin on the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. friedelin 24-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 30122094-3 2018 OBJECTIVE: This study investigates the inhibitory effects of Friedelin on the major human liver CYP isoforms (CYP3A4, 1A2, 2A6, 2E1, 2D6, 2C9, 2C19 and 2C8). friedelin 61-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 30122094-4 2018 MATERIALS AND METHODS: First, the inhibitory effects of Friedelin (100 muM) on the eight human liver CYP isoforms were investigated in vitro using human liver microsomes (HLMs), and then enzyme inhibition, kinetic studies, and time-dependent inhibition studies were conducted to investigate the IC50, Ki and Kinact/KI values of Friedelin. friedelin 56-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 30122094-5 2018 RESULTS: The results indicate that Friedelin inhibited the activity of CYP3A4 and 2E1, with the IC50 values of 10.79 and 22.54 muM, respectively, but other CYP isoforms were not affected. friedelin 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 30122094-8 2018 DISCUSSION AND CONCLUSION: The in vitro studies of Friedelin with CYP isoforms suggested that Friedelin has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP3A4 and 2E1. friedelin 51-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 30122094-8 2018 DISCUSSION AND CONCLUSION: The in vitro studies of Friedelin with CYP isoforms suggested that Friedelin has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP3A4 and 2E1. friedelin 94-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 30285425-6 2018 On a separate note, the cytochrome P450 (CYP) epoxygenase CYP2J2 is the most abundant CYP in the heart where it catalyzes the metabolism of AA and AA-derived eCBs to bioactive epoxides that are involved in diverse cardiovascular functions. ecbs 158-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 30285425-6 2018 On a separate note, the cytochrome P450 (CYP) epoxygenase CYP2J2 is the most abundant CYP in the heart where it catalyzes the metabolism of AA and AA-derived eCBs to bioactive epoxides that are involved in diverse cardiovascular functions. ecbs 158-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 30285425-6 2018 On a separate note, the cytochrome P450 (CYP) epoxygenase CYP2J2 is the most abundant CYP in the heart where it catalyzes the metabolism of AA and AA-derived eCBs to bioactive epoxides that are involved in diverse cardiovascular functions. Epoxy Compounds 176-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 30285425-6 2018 On a separate note, the cytochrome P450 (CYP) epoxygenase CYP2J2 is the most abundant CYP in the heart where it catalyzes the metabolism of AA and AA-derived eCBs to bioactive epoxides that are involved in diverse cardiovascular functions. Epoxy Compounds 176-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 30285425-6 2018 On a separate note, the cytochrome P450 (CYP) epoxygenase CYP2J2 is the most abundant CYP in the heart where it catalyzes the metabolism of AA and AA-derived eCBs to bioactive epoxides that are involved in diverse cardiovascular functions. Epoxy Compounds 176-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 30285425-8 2018 As a result, the role of O-AEA as an endogenous eCB inhibitor of CYP2J2 may provide a new mode of regulation to control the activity of cardiovascular CYP2J2 in vivo and suggests a potential cross-talk between the cardiovascular endocannabinoids and the cytochrome P450 system. virodhamine 25-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 254-269 29752906-5 2018 Various risk factors such as medical comorbidities, use or abuse of supratherapeutic doses of tramadol, and concomitant administration of proconvulsant serotonergic cytochrome P-450 inhibitors will help clinicians identify individuals at an elevated risk for serotonin toxicity and seizures. Serotonin 259-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-181 30501426-2 2018 Different CYP isoenzymes mediate metabolism of many endogenous substrates such as monoaminergic neurotransmitters, neurosteroids, cholesterol, vitamins and arachidonic acid. Cholesterol 130-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-13 30501426-2 2018 Different CYP isoenzymes mediate metabolism of many endogenous substrates such as monoaminergic neurotransmitters, neurosteroids, cholesterol, vitamins and arachidonic acid. Arachidonic Acid 156-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-13 30264400-11 2018 CYP function was determined by measuring resorufin formation and oxcarbazepine (OXC) metabolism. resorufin 41-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 30501426-5 2018 The article focuses on the role of cerebral CYP isoforms in the metabolism of neurotransmitters, neurosteroids, and cholesterol, and their possible involvement in animal behavior, as well as in stress, depression, schizophrenia, cognitive processes, learning, and memory. Cholesterol 116-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 30501426-6 2018 CYP-mediated alternative pathways of dopamine and serotonin synthesis may have a significant role in the local production of these neurotransmitters in the brain regions where the disturbances of these neurotransmitter systems are observed in depression and schizophrenia. Dopamine 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 30501426-6 2018 CYP-mediated alternative pathways of dopamine and serotonin synthesis may have a significant role in the local production of these neurotransmitters in the brain regions where the disturbances of these neurotransmitter systems are observed in depression and schizophrenia. Serotonin 50-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 30264400-11 2018 CYP function was determined by measuring resorufin formation and oxcarbazepine (OXC) metabolism. Oxcarbazepine 65-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 30051214-0 2018 Impact of CYP genotype and inflammatory markers on the plasma concentrations of tramadol and its demethylated metabolites and drug tolerability in cancer patients. Tramadol 80-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-13 30264400-11 2018 CYP function was determined by measuring resorufin formation and oxcarbazepine (OXC) metabolism. Oxcarbazepine 80-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 30051214-2 2018 This study aimed to evaluate the impacts of cytochrome P450 (CYP) genotype and serum inflammatory markers on the plasma concentrations of tramadol and its demethylated metabolites and drug tolerability in cancer patients. Tramadol 138-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 30264400-14 2018 GR EPI-EC silencing or inhibition led to (1) increased Pphenytoin BBB permeability as compared to control; (2) decreased CYP function, indirectly evaluated by resorufin formation; (3) improved OXC bioavailability with increased abluminal (brain-side) OXC levels as compared to control. epi-ec 3-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 30051214-2 2018 This study aimed to evaluate the impacts of cytochrome P450 (CYP) genotype and serum inflammatory markers on the plasma concentrations of tramadol and its demethylated metabolites and drug tolerability in cancer patients. Tramadol 138-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 30373287-0 2018 beta-Naphtoflavone and Ethanol Induce Cytochrome P450 and Protect towards MPP+ Toxicity in Human Neuroblastoma SH-SY5Y Cells. beta-naphtoflavone 0-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 30253245-4 2018 The effects of 2-HOBA on the mRNA expression of select cytochrome P450 (CYP) enzymes were also assessed in cryopreserved human hepatocytes. 2-(aminomethyl)phenol 15-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 30373287-0 2018 beta-Naphtoflavone and Ethanol Induce Cytochrome P450 and Protect towards MPP+ Toxicity in Human Neuroblastoma SH-SY5Y Cells. Ethanol 23-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 30373287-4 2018 We have studied the inducibility of CYP isozymes in human neuroblastoma SH-SY5Y cells, treated with beta-naphtoflavone (beta-NF) or ethanol (EtOH) as inducers, by qRT-PCR, Western blot (WB), and metabolic activity assays. beta-naphtoflavone 100-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 30373287-4 2018 We have studied the inducibility of CYP isozymes in human neuroblastoma SH-SY5Y cells, treated with beta-naphtoflavone (beta-NF) or ethanol (EtOH) as inducers, by qRT-PCR, Western blot (WB), and metabolic activity assays. beta-Naphthoflavone 120-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 30373287-4 2018 We have studied the inducibility of CYP isozymes in human neuroblastoma SH-SY5Y cells, treated with beta-naphtoflavone (beta-NF) or ethanol (EtOH) as inducers, by qRT-PCR, Western blot (WB), and metabolic activity assays. Ethanol 132-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 30373287-4 2018 We have studied the inducibility of CYP isozymes in human neuroblastoma SH-SY5Y cells, treated with beta-naphtoflavone (beta-NF) or ethanol (EtOH) as inducers, by qRT-PCR, Western blot (WB), and metabolic activity assays. Ethanol 141-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 30109772-1 2018 An organic semiconducting polymer nanobiocatalyst (SPNB) composed of a semiconducting polymer core conjugated with microsomal cytochrome P450 (CYP) has been developed for photoactivation of intracellular redox. Polymers 26-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-141 30322383-2 2018 In addition to nuclear receptors, there are other intracellular RA binding proteins such as cellular retinoic acid binding proteins (CRABP1 and CRABP2) and cytochrome P450 (CYP) enzymes, whose contributions to the RA signaling pathway have not been fully understood. Tretinoin 64-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-171 30322383-2 2018 In addition to nuclear receptors, there are other intracellular RA binding proteins such as cellular retinoic acid binding proteins (CRABP1 and CRABP2) and cytochrome P450 (CYP) enzymes, whose contributions to the RA signaling pathway have not been fully understood. Tretinoin 64-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-176 30322383-2 2018 In addition to nuclear receptors, there are other intracellular RA binding proteins such as cellular retinoic acid binding proteins (CRABP1 and CRABP2) and cytochrome P450 (CYP) enzymes, whose contributions to the RA signaling pathway have not been fully understood. Tretinoin 134-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-176 30109772-1 2018 An organic semiconducting polymer nanobiocatalyst (SPNB) composed of a semiconducting polymer core conjugated with microsomal cytochrome P450 (CYP) has been developed for photoactivation of intracellular redox. Polymers 26-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-146 30109772-2 2018 The core serves as the light-harvesting unit to initiate photoinduced electron transfer (PET) and facilitate the regeneration of dihydronicotinamide adenine dinucleotide phosphate (NADPH), while CYP is the catalytic center for intracellular redox. NADP 181-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-198 30073515-1 2018 We report the development and characterization of digital microfluidic (DMF) immobilized enzyme reactors (IMERs) for studying cytochrome P450 (CYP)-mediated drug metabolism on droplet scale. Dimethylformamide 72-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-141 30073515-1 2018 We report the development and characterization of digital microfluidic (DMF) immobilized enzyme reactors (IMERs) for studying cytochrome P450 (CYP)-mediated drug metabolism on droplet scale. Dimethylformamide 72-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-146 30073515-3 2018 The DMF devices also incorporate inexpensive, inkjet-printed microheaters for on-demand regio-specific heating of the IMERs to physiological temperature, which is crucial for maintaining the activity of the temperature-sensitive CYP reaction. Dimethylformamide 4-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 229-232 29858511-3 2018 Clinical relevance of the cytochrome P450 (CYP) polymorphism related to dose, effectiveness and/or toxicity of key drugs are presented in this review, including that of warfarin, clopidogrel, tricyclic antidepressants, and proton pump inhibitors. Warfarin 169-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 29858511-3 2018 Clinical relevance of the cytochrome P450 (CYP) polymorphism related to dose, effectiveness and/or toxicity of key drugs are presented in this review, including that of warfarin, clopidogrel, tricyclic antidepressants, and proton pump inhibitors. Warfarin 169-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 29858511-3 2018 Clinical relevance of the cytochrome P450 (CYP) polymorphism related to dose, effectiveness and/or toxicity of key drugs are presented in this review, including that of warfarin, clopidogrel, tricyclic antidepressants, and proton pump inhibitors. Clopidogrel 179-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 29858511-3 2018 Clinical relevance of the cytochrome P450 (CYP) polymorphism related to dose, effectiveness and/or toxicity of key drugs are presented in this review, including that of warfarin, clopidogrel, tricyclic antidepressants, and proton pump inhibitors. Clopidogrel 179-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 30262788-5 2018 In this review, an overview will be given on the therapeutically most important classes of pharmacokinetic enhancers like beta-lactamase inhibitors, inhibitors of CYP (cytochrome P450) enzymes in HIV therapy and hepatitis C, boosters for fluoropyrimidine-type anticancer agents, compounds utilized for enabling therapy of Parkinson"s disease with levodopa, and others. 2-fluoropyrimidine 238-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-183 29762875-2 2018 In vitro data showed that pomalidomide is a substrate of multiple cytochrome P450 (CYP) isozymes and that its oxidative metabolism is mediated primarily by CYP1A2 and CYP3A4, with minor contributions from CYP2C19 and CYP2D6. pomalidomide 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 29762875-2 2018 In vitro data showed that pomalidomide is a substrate of multiple cytochrome P450 (CYP) isozymes and that its oxidative metabolism is mediated primarily by CYP1A2 and CYP3A4, with minor contributions from CYP2C19 and CYP2D6. pomalidomide 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 30262788-5 2018 In this review, an overview will be given on the therapeutically most important classes of pharmacokinetic enhancers like beta-lactamase inhibitors, inhibitors of CYP (cytochrome P450) enzymes in HIV therapy and hepatitis C, boosters for fluoropyrimidine-type anticancer agents, compounds utilized for enabling therapy of Parkinson"s disease with levodopa, and others. Levodopa 347-355 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-183 30066055-1 2018 While cytochrome P450 (CYP)-mediated biosynthesis of arachidonic acid (AA) epoxides promotes tumor growth by driving angiogenesis, cancer cell intrinsic functions of CYPs are less understood. arachidonic acid (aa) epoxides 53-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 29858741-3 2018 Eicosanoids, which are metabolites of polyunsaturated fatty acids produced by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes, are important lipid-signaling molecules involved in the regulation of inflammation and tumorigenesis. Eicosanoids 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 29858741-3 2018 Eicosanoids, which are metabolites of polyunsaturated fatty acids produced by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes, are important lipid-signaling molecules involved in the regulation of inflammation and tumorigenesis. Eicosanoids 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-144 29858741-3 2018 Eicosanoids, which are metabolites of polyunsaturated fatty acids produced by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes, are important lipid-signaling molecules involved in the regulation of inflammation and tumorigenesis. Fatty Acids, Unsaturated 38-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 29858741-3 2018 Eicosanoids, which are metabolites of polyunsaturated fatty acids produced by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes, are important lipid-signaling molecules involved in the regulation of inflammation and tumorigenesis. Fatty Acids, Unsaturated 38-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-144 30066055-1 2018 While cytochrome P450 (CYP)-mediated biosynthesis of arachidonic acid (AA) epoxides promotes tumor growth by driving angiogenesis, cancer cell intrinsic functions of CYPs are less understood. arachidonic acid (aa) epoxides 53-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 30066055-2 2018 CYP-derived AA epoxides, called epoxyeicosatrienoic acids (EETs), also promote the growth of tumor epithelia. epoxyeicosatrienoic acids 32-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 30066055-2 2018 CYP-derived AA epoxides, called epoxyeicosatrienoic acids (EETs), also promote the growth of tumor epithelia. eets 59-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29226313-0 2018 Model-Based Assessments of CYP-Mediated Drug-Drug Interaction Risk of Alectinib: Physiologically Based Pharmacokinetic Modeling Supported Clinical Development. alectinib 70-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 29226313-2 2018 Alectinib and its major active metabolite M4 exhibited drug-drug interaction (DDI) potential through cytochrome P450 (CYP) enzymes CYP3A4 and CYP2C8 in vitro. alectinib 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 29226313-6 2018 This work supports that alectinib can be prescribed without dose adjustment for CYP-mediated DDI liabilities. alectinib 24-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 30066055-4 2018 Recently, the diabetes drug metformin was found to inhibit CYP AA epoxygenase activity, allowing the design of more potent biguanides to target tumor growth. Metformin 28-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 29226313-2 2018 Alectinib and its major active metabolite M4 exhibited drug-drug interaction (DDI) potential through cytochrome P450 (CYP) enzymes CYP3A4 and CYP2C8 in vitro. alectinib 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 30066055-4 2018 Recently, the diabetes drug metformin was found to inhibit CYP AA epoxygenase activity, allowing the design of more potent biguanides to target tumor growth. Biguanides 123-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 30278485-1 2018 BACKGROUND: Human cytochrome P450 (CYP) is an enzyme responsible for the metabolic activation of many carcinogens, including nitrosamines. Nitrosamines 125-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 29496531-7 2018 The results suggested that multiple dose administration of Danshen capsules could induce cytochrome P450 (CYP) isoenzymes, thereby increasing the clearance of clopidogrel. Clopidogrel 159-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 29496531-7 2018 The results suggested that multiple dose administration of Danshen capsules could induce cytochrome P450 (CYP) isoenzymes, thereby increasing the clearance of clopidogrel. Clopidogrel 159-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 30170758-2 2018 The primary enzyme involved in the metabolism of ertugliflozin is uridine diphosphate-glucuronosyltransferase (UGT) 1A9, with minor contributions from UGT2B7 and cytochrome P450 (CYP) isoenzymes 3A4, 3A5, and 2C8. ertugliflozin 49-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 30170758-2 2018 The primary enzyme involved in the metabolism of ertugliflozin is uridine diphosphate-glucuronosyltransferase (UGT) 1A9, with minor contributions from UGT2B7 and cytochrome P450 (CYP) isoenzymes 3A4, 3A5, and 2C8. ertugliflozin 49-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-182 30429697-3 2018 Triazole antifungals are known inhibitors of cytochrome P450 (CYP) enzymes. Triazoles 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 30429697-3 2018 Triazole antifungals are known inhibitors of cytochrome P450 (CYP) enzymes. Triazoles 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 30278485-1 2018 BACKGROUND: Human cytochrome P450 (CYP) is an enzyme responsible for the metabolic activation of many carcinogens, including nitrosamines. Nitrosamines 125-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 30121710-3 2018 Paracetamol is known to be metabolized into N-(4-hydroxyphenyl)-arachidonamide (AM404) via fatty acid amide hydrolase (FAAH) and into N-acetyl-p-benzoquinone imine (NAPQI) via cytochrome P450 (CYP) enzymes. Acetaminophen 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-191 30200214-5 2018 In this study, we elucidated the structure of generated metabolites using a high-resolution quadrupole-orbitrap mass spectrometer (HR-MS/MS) and characterized the major human cytochrome P450 (CYP) and uridine 5"-diphospho-glucuronosyltransferase (UGT) isozymes involved in osthenol metabolism in human liver microsomes (HLMs). osthenol 273-281 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-190 30200214-5 2018 In this study, we elucidated the structure of generated metabolites using a high-resolution quadrupole-orbitrap mass spectrometer (HR-MS/MS) and characterized the major human cytochrome P450 (CYP) and uridine 5"-diphospho-glucuronosyltransferase (UGT) isozymes involved in osthenol metabolism in human liver microsomes (HLMs). osthenol 273-281 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-195 30052045-0 2018 Proximal Pocket Controls Alkene Oxidation Selectivity of Cytochrome P450 and Chloroperoxidase toward Small, Nonpolar Substrates. Alkenes 25-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-93 30121710-3 2018 Paracetamol is known to be metabolized into N-(4-hydroxyphenyl)-arachidonamide (AM404) via fatty acid amide hydrolase (FAAH) and into N-acetyl-p-benzoquinone imine (NAPQI) via cytochrome P450 (CYP) enzymes. Acetaminophen 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-196 30121710-3 2018 Paracetamol is known to be metabolized into N-(4-hydroxyphenyl)-arachidonamide (AM404) via fatty acid amide hydrolase (FAAH) and into N-acetyl-p-benzoquinone imine (NAPQI) via cytochrome P450 (CYP) enzymes. N-(4-hydroxyphenyl)arachidonylamide 80-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-191 30121710-3 2018 Paracetamol is known to be metabolized into N-(4-hydroxyphenyl)-arachidonamide (AM404) via fatty acid amide hydrolase (FAAH) and into N-acetyl-p-benzoquinone imine (NAPQI) via cytochrome P450 (CYP) enzymes. N-(4-hydroxyphenyl)arachidonylamide 80-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-196 30121710-3 2018 Paracetamol is known to be metabolized into N-(4-hydroxyphenyl)-arachidonamide (AM404) via fatty acid amide hydrolase (FAAH) and into N-acetyl-p-benzoquinone imine (NAPQI) via cytochrome P450 (CYP) enzymes. N-acetyl-4-benzoquinoneimine 134-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-191 30121710-3 2018 Paracetamol is known to be metabolized into N-(4-hydroxyphenyl)-arachidonamide (AM404) via fatty acid amide hydrolase (FAAH) and into N-acetyl-p-benzoquinone imine (NAPQI) via cytochrome P450 (CYP) enzymes. N-acetyl-4-benzoquinoneimine 134-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-196 30121710-5 2018 In addition, paracetamol has the potential to interact with other drugs that are also involved with CYP family enzymes (inducer/inhibitor/substrate), an example being illicit drugs. Acetaminophen 13-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 30072626-0 2018 Exploring the Metabolism of Loxoprofen in Liver Microsomes: The Role of Cytochrome P450 and UDP-Glucuronosyltransferase in Its Biotransformation. loxoprofen 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 30072626-4 2018 In the present study, we investigated metabolic enzymes, such as cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT), which are involved in the metabolism of loxoprofen. loxoprofen 166-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 30072626-4 2018 In the present study, we investigated metabolic enzymes, such as cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT), which are involved in the metabolism of loxoprofen. loxoprofen 166-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 30072626-6 2018 Based on the results for the formation of metabolites when incubated in dexamethasone-induced microsomes, incubation with ketoconazole, and human recombinant cDNA-expressed cytochrome P450s, we identified CYP3A4 and CYP3A5 as the major CYP isoforms involved in the hydroxylation of loxoprofen (M3 and M4). Dexamethasone 72-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-208 30072626-6 2018 Based on the results for the formation of metabolites when incubated in dexamethasone-induced microsomes, incubation with ketoconazole, and human recombinant cDNA-expressed cytochrome P450s, we identified CYP3A4 and CYP3A5 as the major CYP isoforms involved in the hydroxylation of loxoprofen (M3 and M4). Ketoconazole 122-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-208 29908721-1 2018 CYP2C and CYP2 J enzymes, commonly named as cytochrome P450 (CYP) epoxygenases, convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs), biologically active eicosanoids with many functions in organism. eets 154-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29908721-1 2018 CYP2C and CYP2 J enzymes, commonly named as cytochrome P450 (CYP) epoxygenases, convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs), biologically active eicosanoids with many functions in organism. Eicosanoids 181-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 29908721-1 2018 CYP2C and CYP2 J enzymes, commonly named as cytochrome P450 (CYP) epoxygenases, convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs), biologically active eicosanoids with many functions in organism. Arachidonic Acid 88-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 29908721-1 2018 CYP2C and CYP2 J enzymes, commonly named as cytochrome P450 (CYP) epoxygenases, convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs), biologically active eicosanoids with many functions in organism. Arachidonic Acid 88-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 30072626-6 2018 Based on the results for the formation of metabolites when incubated in dexamethasone-induced microsomes, incubation with ketoconazole, and human recombinant cDNA-expressed cytochrome P450s, we identified CYP3A4 and CYP3A5 as the major CYP isoforms involved in the hydroxylation of loxoprofen (M3 and M4). loxoprofen 282-292 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-208 29908721-1 2018 CYP2C and CYP2 J enzymes, commonly named as cytochrome P450 (CYP) epoxygenases, convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs), biologically active eicosanoids with many functions in organism. epoxyeicosatrienoic acids 127-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 29908721-1 2018 CYP2C and CYP2 J enzymes, commonly named as cytochrome P450 (CYP) epoxygenases, convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs), biologically active eicosanoids with many functions in organism. Eicosanoids 181-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29908721-11 2018 As the placenta is absolutely crucial for prenatal development, arachidonic acid is essential part of human nutrient and CYP epoxygenases expression can be affected by xenobiotics, further investigation of the exact role of CYP epoxygenases, sEH, and their metabolites in normal pregnancy and under pathological conditions is needed. Arachidonic Acid 64-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 29908721-1 2018 CYP2C and CYP2 J enzymes, commonly named as cytochrome P450 (CYP) epoxygenases, convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs), biologically active eicosanoids with many functions in organism. epoxyeicosatrienoic acids 127-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29027195-3 2018 Here we show that TSPYLs, especially TSPYL 1, 2, and 4, can regulate the expression of many CYP genes, including CYP17A1, a key enzyme in androgen biosynthesis, and CYP3A4, an enzyme that catalyzes the metabolism of abiraterone, a CYP17 inhibitor. abiraterone 216-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 29908721-1 2018 CYP2C and CYP2 J enzymes, commonly named as cytochrome P450 (CYP) epoxygenases, convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs), biologically active eicosanoids with many functions in organism. eets 154-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 29916051-4 2018 This review collects available information of which-taken with great caution because of the still very limited data-the most salient points are: in the skin of all animal species and skin-derived in vitro systems considered in this review cytochrome P450 (CYP)-dependent monooxygenase activities (largely responsible for initiating xenobiotica metabolism in the organ which provides most of the xenobiotica metabolism of the mammalian organism, the liver) are very low to undetectable. xenobiotica 332-343 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 239-254 29916051-4 2018 This review collects available information of which-taken with great caution because of the still very limited data-the most salient points are: in the skin of all animal species and skin-derived in vitro systems considered in this review cytochrome P450 (CYP)-dependent monooxygenase activities (largely responsible for initiating xenobiotica metabolism in the organ which provides most of the xenobiotica metabolism of the mammalian organism, the liver) are very low to undetectable. xenobiotica 332-343 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 256-259 29916051-6 2018 Moreover, conjugating enzyme activities such as glutathione transferases and glucuronosyltransferases are much higher than the oxidative CYP activities. Glutathione 48-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 29722926-4 2018 Akin to cytochrome-b5 (cyt-b5 ), Arg 125 on the C-helix of CYP450s is found to be important for effective electron transfer, thus supporting the competitive behavior of redox partners for CYP450s. Arginine 33-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-65 30039377-6 2018 This review describes the metabolic pathways of 13 prevalent synthetic cannabinoids and various drug-metabolizing enzymes responsible for their metabolism, including cytochrome P450 (CYP), UDP-glucuronosyltransferases (UGTs), and carboxylesterases. Cannabinoids 71-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-181 30039377-6 2018 This review describes the metabolic pathways of 13 prevalent synthetic cannabinoids and various drug-metabolizing enzymes responsible for their metabolism, including cytochrome P450 (CYP), UDP-glucuronosyltransferases (UGTs), and carboxylesterases. Cannabinoids 71-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-186 30039377-7 2018 The inhibitory effects of synthetic cannabinoids on CYP and UGT activities are also reviewed to predict the potential of synthetic cannabinoids for drug-drug interactions. Cannabinoids 36-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 30039377-8 2018 The drug-metabolizing enzymes responsible for metabolism of synthetic cannabinoids should be characterized and the effects of synthetic cannabinoids on CYP and UGT activities should be determined to predict the pharmacokinetics of synthetic cannabinoids and synthetic cannabinoid-induced drug-drug interactions in the clinic. Cannabinoids 136-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-155 30039377-8 2018 The drug-metabolizing enzymes responsible for metabolism of synthetic cannabinoids should be characterized and the effects of synthetic cannabinoids on CYP and UGT activities should be determined to predict the pharmacokinetics of synthetic cannabinoids and synthetic cannabinoid-induced drug-drug interactions in the clinic. Cannabinoids 136-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-155 29921510-0 2018 The regulation mechanism of AhR activated by benzo[a]pyrene for CYP expression are different between 2D and 3D culture of human lung cancer cells. Benzo(a)pyrene 45-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 29751197-0 2018 Investigating Cytochrome P450 specificity during glycopeptide antibiotic biosynthesis through a homologue hybridization approach. Glycopeptides 49-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 29746911-3 2018 Previous studies suggested that 4-MAA demethylation can be performed by hepatic cytochrome P450 (CYP) 3A4, but the possible contribution of other CYPs remains unclear. 4-maa 32-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 29746911-3 2018 Previous studies suggested that 4-MAA demethylation can be performed by hepatic cytochrome P450 (CYP) 3A4, but the possible contribution of other CYPs remains unclear. 4-maa 32-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 29746911-5 2018 Based on CYP induction (HepaRG cells) and CYP inhibition (HLM) we could identify CYP2B6, 2C8, 2C9 and 3A4 as major contributors to 4-MAA demethylation. 4-maa 131-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 29746911-5 2018 Based on CYP induction (HepaRG cells) and CYP inhibition (HLM) we could identify CYP2B6, 2C8, 2C9 and 3A4 as major contributors to 4-MAA demethylation. 4-maa 131-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 29458047-0 2018 Effects of cytochrome P450 single nucleotide polymorphisms on methadone metabolism and pharmacodynamics. Methadone 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-26 29458047-4 2018 Methadone is primarily metabolized in the liver by cytochrome P450 (CYP) enzymes, predominately by CYP2B6, followed by CYP3A4, 2C19, 2D6, and to a lesser extent, CYP2C18, 3A7, 2C8, 2C9, 3A5, and 1A2. Methadone 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-66 29458047-4 2018 Methadone is primarily metabolized in the liver by cytochrome P450 (CYP) enzymes, predominately by CYP2B6, followed by CYP3A4, 2C19, 2D6, and to a lesser extent, CYP2C18, 3A7, 2C8, 2C9, 3A5, and 1A2. Methadone 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 29458047-9 2018 The involvement, contribution, and understanding the role of SNPs in CYP2B6, and other CYP genes, in methadone metabolism can improve the therapeutic uses of methadone in patient outcome and the development of personalized medicine. Methadone 101-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 29458047-9 2018 The involvement, contribution, and understanding the role of SNPs in CYP2B6, and other CYP genes, in methadone metabolism can improve the therapeutic uses of methadone in patient outcome and the development of personalized medicine. Methadone 158-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 29888644-0 2018 Regulation of cytochrome P450 gene expression by ketamine: a review. Ketamine 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 29735753-0 2018 Metabolism and Disposition of Siponimod, a Novel Selective S1P1/S1P5 Agonist, in Healthy Volunteers and In Vitro Identification of Human Cytochrome P450 Enzymes Involved in Its Oxidative Metabolism. siponimod 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-152 29888644-2 2018 Ketamine undergoes extensive oxidative metabolism by cytochrome P450 (CYP) enzymes. Ketamine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 29888644-2 2018 Ketamine undergoes extensive oxidative metabolism by cytochrome P450 (CYP) enzymes. Ketamine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 29888644-3 2018 Considerable efforts have been expended to elucidate the ketamine-induced regulation of CYP gene expression. Ketamine 57-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 29888644-5 2018 Understanding how ketamine regulates CYP gene expression is clinically meaningful. Ketamine 18-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 29888644-6 2018 Areas covered: In this article, the authors provide a brief review of clinical applications of ketamine and its metabolism by CYP enzymes. Ketamine 95-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 29888644-7 2018 We discuss the effects of ketamine on the regulation of CYP gene expression, exploring aspects of cytoskeletal remodeling, mitochondrial functions, and calcium homeostasis. Ketamine 26-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 29888644-8 2018 Expert opinion: Ketamine may inhibit CYP gene expression through inhibiting calcium signaling, decreasing ATP levels, producing excessive reactive oxygen species, and subsequently perturbing cytoskeletal dynamics. Ketamine 16-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 29888644-8 2018 Expert opinion: Ketamine may inhibit CYP gene expression through inhibiting calcium signaling, decreasing ATP levels, producing excessive reactive oxygen species, and subsequently perturbing cytoskeletal dynamics. Calcium 76-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 29719203-0 2018 The burgeoning role of cytochrome P450-mediated vitamin D metabolites against colorectal cancer. Vitamin D 48-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 29719203-1 2018 The metabolites of vitamin D3 (VD3) mediated by different cytochrome P450 (CYP) enzymes, play fundamental roles in many physiological processes in relation to human health. Cholecalciferol 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 29888644-8 2018 Expert opinion: Ketamine may inhibit CYP gene expression through inhibiting calcium signaling, decreasing ATP levels, producing excessive reactive oxygen species, and subsequently perturbing cytoskeletal dynamics. Adenosine Triphosphate 106-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 29719203-1 2018 The metabolites of vitamin D3 (VD3) mediated by different cytochrome P450 (CYP) enzymes, play fundamental roles in many physiological processes in relation to human health. Cholecalciferol 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 29888644-8 2018 Expert opinion: Ketamine may inhibit CYP gene expression through inhibiting calcium signaling, decreasing ATP levels, producing excessive reactive oxygen species, and subsequently perturbing cytoskeletal dynamics. Reactive Oxygen Species 138-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 28686070-0 2018 Effects of aging and rifampicin pretreatment on the pharmacokinetics of human cytochrome P450 probes caffeine, warfarin, omeprazole, metoprolol and midazolam in common marmosets genotyped for cytochrome P450 2C19. Rifampin 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 29989011-0 2018 Inhibition of cytochrome P450 enzymes by thymoquinone in human liver microsomes. thymoquinone 41-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 29989011-2 2018 The inhibition of CYP enzymatic activities by TQ was evaluated by incubating typical substrates (phenacetin for CYP1A2, tolbutamide for CYP2C9, dextromethorphan for CYP2D6, and testosterone for CYP3A4) with human liver microsomes and NADPH in the absence or presence of TQ (1, 10 and 100 microM). thymoquinone 46-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 29989011-2 2018 The inhibition of CYP enzymatic activities by TQ was evaluated by incubating typical substrates (phenacetin for CYP1A2, tolbutamide for CYP2C9, dextromethorphan for CYP2D6, and testosterone for CYP3A4) with human liver microsomes and NADPH in the absence or presence of TQ (1, 10 and 100 microM). Phenacetin 97-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 29989011-2 2018 The inhibition of CYP enzymatic activities by TQ was evaluated by incubating typical substrates (phenacetin for CYP1A2, tolbutamide for CYP2C9, dextromethorphan for CYP2D6, and testosterone for CYP3A4) with human liver microsomes and NADPH in the absence or presence of TQ (1, 10 and 100 microM). Tolbutamide 120-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 29989011-2 2018 The inhibition of CYP enzymatic activities by TQ was evaluated by incubating typical substrates (phenacetin for CYP1A2, tolbutamide for CYP2C9, dextromethorphan for CYP2D6, and testosterone for CYP3A4) with human liver microsomes and NADPH in the absence or presence of TQ (1, 10 and 100 microM). Dextromethorphan 144-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 29989011-2 2018 The inhibition of CYP enzymatic activities by TQ was evaluated by incubating typical substrates (phenacetin for CYP1A2, tolbutamide for CYP2C9, dextromethorphan for CYP2D6, and testosterone for CYP3A4) with human liver microsomes and NADPH in the absence or presence of TQ (1, 10 and 100 microM). Testosterone 177-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 29989011-2 2018 The inhibition of CYP enzymatic activities by TQ was evaluated by incubating typical substrates (phenacetin for CYP1A2, tolbutamide for CYP2C9, dextromethorphan for CYP2D6, and testosterone for CYP3A4) with human liver microsomes and NADPH in the absence or presence of TQ (1, 10 and 100 microM). NADP 234-239 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 29989011-2 2018 The inhibition of CYP enzymatic activities by TQ was evaluated by incubating typical substrates (phenacetin for CYP1A2, tolbutamide for CYP2C9, dextromethorphan for CYP2D6, and testosterone for CYP3A4) with human liver microsomes and NADPH in the absence or presence of TQ (1, 10 and 100 microM). thymoquinone 270-272 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 28686070-0 2018 Effects of aging and rifampicin pretreatment on the pharmacokinetics of human cytochrome P450 probes caffeine, warfarin, omeprazole, metoprolol and midazolam in common marmosets genotyped for cytochrome P450 2C19. Caffeine 101-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 28686070-0 2018 Effects of aging and rifampicin pretreatment on the pharmacokinetics of human cytochrome P450 probes caffeine, warfarin, omeprazole, metoprolol and midazolam in common marmosets genotyped for cytochrome P450 2C19. Warfarin 111-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 28686070-0 2018 Effects of aging and rifampicin pretreatment on the pharmacokinetics of human cytochrome P450 probes caffeine, warfarin, omeprazole, metoprolol and midazolam in common marmosets genotyped for cytochrome P450 2C19. Omeprazole 121-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 28686070-0 2018 Effects of aging and rifampicin pretreatment on the pharmacokinetics of human cytochrome P450 probes caffeine, warfarin, omeprazole, metoprolol and midazolam in common marmosets genotyped for cytochrome P450 2C19. Metoprolol 133-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 28686070-0 2018 Effects of aging and rifampicin pretreatment on the pharmacokinetics of human cytochrome P450 probes caffeine, warfarin, omeprazole, metoprolol and midazolam in common marmosets genotyped for cytochrome P450 2C19. Midazolam 148-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 28686070-2 2018 The pharmacokinetics were investigated for human cytochrome P450 probes after single intravenous and oral administrations of 0.20 and 1.0 mg/kg, respectively, of caffeine, warfarin, omeprazole, metoprolol and midazolam to aged (10-14 years old, n = 4) or rifampicin-treated/young (3 years old, n = 3) male common marmosets all genotyped as heterozygous for a cytochrome P450 2C19 variant. Caffeine 162-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 29759551-10 2018 Additionally, glucocorticoids, asparaginase, anthracycline, vincristine and cytarabine that trans-repress gene expression, deprives cells of asparagine, triggers cell cycle arrest, influences cytochrome-P450 polymorphism and inhibits DNA polymerase, respectively, have been used in chemotherapy in ALL patients. Anthracyclines 45-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-207 29914111-6 2018 For example, cytochrome P450 enzymes that metabolize statins may affect n-3 long chain polyunsaturated fatty acid metabolism and vice versa. n-3 long chain polyunsaturated fatty acid 72-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 29196978-1 2018 Several studies have demonstrated the role of cytochrome P450 (CYP) and its associated arachidonic acid (AA) metabolites in the anthracyclines-induced cardiac toxicity. Arachidonic Acid 87-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 29196978-1 2018 Several studies have demonstrated the role of cytochrome P450 (CYP) and its associated arachidonic acid (AA) metabolites in the anthracyclines-induced cardiac toxicity. Arachidonic Acid 87-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 29196978-1 2018 Several studies have demonstrated the role of cytochrome P450 (CYP) and its associated arachidonic acid (AA) metabolites in the anthracyclines-induced cardiac toxicity. Anthracyclines 128-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 29196978-1 2018 Several studies have demonstrated the role of cytochrome P450 (CYP) and its associated arachidonic acid (AA) metabolites in the anthracyclines-induced cardiac toxicity. Anthracyclines 128-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 29196978-2 2018 However, the ability of daunorubicin (DNR) to induce cardiotoxicity through the modulation of CYP and its associated AA metabolites has not been investigated yet. Daunorubicin 24-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 29621538-0 2018 Ketamine and ketamine metabolites as novel estrogen receptor ligands: Induction of cytochrome P450 and AMPA glutamate receptor gene expression. Ketamine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-98 29621538-0 2018 Ketamine and ketamine metabolites as novel estrogen receptor ligands: Induction of cytochrome P450 and AMPA glutamate receptor gene expression. Ketamine 13-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-98 29759551-10 2018 Additionally, glucocorticoids, asparaginase, anthracycline, vincristine and cytarabine that trans-repress gene expression, deprives cells of asparagine, triggers cell cycle arrest, influences cytochrome-P450 polymorphism and inhibits DNA polymerase, respectively, have been used in chemotherapy in ALL patients. Vincristine 60-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-207 29759551-10 2018 Additionally, glucocorticoids, asparaginase, anthracycline, vincristine and cytarabine that trans-repress gene expression, deprives cells of asparagine, triggers cell cycle arrest, influences cytochrome-P450 polymorphism and inhibits DNA polymerase, respectively, have been used in chemotherapy in ALL patients. Cytarabine 76-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-207 29759551-10 2018 Additionally, glucocorticoids, asparaginase, anthracycline, vincristine and cytarabine that trans-repress gene expression, deprives cells of asparagine, triggers cell cycle arrest, influences cytochrome-P450 polymorphism and inhibits DNA polymerase, respectively, have been used in chemotherapy in ALL patients. Asparagine 141-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-207 29650790-0 2018 Effect of Endocannabinoid Oleamide on Rat and Human Liver Cytochrome P450 Enzymes in In Vitro and In Vivo Models. endocannabinoid oleamide 10-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 29576510-0 2018 Probing cytochrome P450 bioactivation and fluorescent properties with morpholinyl-tethered anthraquinones. morpholinyl-tethered anthraquinones 70-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 28639016-8 2018 On the other hand, it was inferred that CB156 is difficult to be metabolized by these four CYP isozymes. cb156 40-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 29264831-0 2018 CYP Suppression in Human Hepatocytes by Monomethyl Auristatin E, the Payload in Brentuximab Vedotin (Adcetris ), is Associated with Microtubule Disruption. monomethyl auristatin E 40-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29526504-0 2018 Efficient hydroxylation of cycloalkanes by co-addition of decoy molecules to variants of the cytochrome P450 CYP102A1. Cycloparaffins 27-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-108 29526504-1 2018 The wild-type cytochrome P450 (CYP) monooxygenase enzyme CYP102A1 (P450Bm3) has low activity for cycloalkane oxidation. Cycloparaffins 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 29526504-1 2018 The wild-type cytochrome P450 (CYP) monooxygenase enzyme CYP102A1 (P450Bm3) has low activity for cycloalkane oxidation. Cycloparaffins 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 28730856-0 2018 In vitro drug-drug interactions of budesonide: inhibition and induction of transporters and cytochrome P450 enzymes. Budesonide 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 28730856-4 2018 This study aimed to evaluate the potential of budesonide to act as a perpetrator or a victim of transporter- or CYP-mediated drug-drug interactions (DDIs). Budesonide 46-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-115 29517843-0 2018 Diverging Mechanisms: Cytochrome-P450-Catalyzed Demethylation and gamma-Lactone Formation in Bacterial Gibberellin Biosynthesis. 4-hexanolide 66-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 29517843-0 2018 Diverging Mechanisms: Cytochrome-P450-Catalyzed Demethylation and gamma-Lactone Formation in Bacterial Gibberellin Biosynthesis. Gibberellins 103-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 29274758-2 2018 The DOACs are P-glycoprotein (P-gp) and cytochrome p-450 (CYP3A4) substrates. doacs 4-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 29530595-1 2018 Cytochrome P450 (CYP) monoxygenses utilize heme cofactors to catalyze oxidation reactions. Heme 43-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29530595-1 2018 Cytochrome P450 (CYP) monoxygenses utilize heme cofactors to catalyze oxidation reactions. Heme 43-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 29530595-9 2018 These results suggest that water-bridged complexes are under-represented in CYP structural databases and can have energies similar to other ligation modes. Water 27-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 29449127-1 2018 BACKGROUND AND GOAL: Cytochrome P450 (CYP) enzymes are responsible for the conversion of clopidogrel into its active metabolite and the metabolism of proton pump inhibitors (PPIs), which may also inhibit CYP enzymes. Clopidogrel 89-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 29449127-1 2018 BACKGROUND AND GOAL: Cytochrome P450 (CYP) enzymes are responsible for the conversion of clopidogrel into its active metabolite and the metabolism of proton pump inhibitors (PPIs), which may also inhibit CYP enzymes. Clopidogrel 89-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 29449127-1 2018 BACKGROUND AND GOAL: Cytochrome P450 (CYP) enzymes are responsible for the conversion of clopidogrel into its active metabolite and the metabolism of proton pump inhibitors (PPIs), which may also inhibit CYP enzymes. Clopidogrel 89-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-207 28657402-0 2018 In vitro evaluation of the inhibition and induction potential of olaparib, a potent poly(ADP-ribose) polymerase inhibitor, on cytochrome P450. olaparib 65-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-141 28657402-2 2018 In vitro studies were conducted to evaluate potential inhibitory and inductive effects of the poly(ADP-ribose) polymerase (PARP) inhibitor, olaparib, on cytochrome P450 (CYP) enzymes. olaparib 140-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-168 28657402-2 2018 In vitro studies were conducted to evaluate potential inhibitory and inductive effects of the poly(ADP-ribose) polymerase (PARP) inhibitor, olaparib, on cytochrome P450 (CYP) enzymes. olaparib 140-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-173 28657402-9 2018 Assessment of the CYP induction potential of olaparib (0.061-44 muM) showed minor concentration-related increases in CYP1A2 and more marked increases in CYP2B6 and CYP3A4 mRNA, compared with positive control activity; however, no significant change in CYP3A4/5 enzyme activity was observed. olaparib 45-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 29719052-0 2018 Effect of single-walled carbon nanotubes on cytochrome P450 activity in human liver microsomes in vitro. Carbon 24-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 29576510-1 2018 Structural features from the anticancer prodrug nemorubicin (MMDX) and the DNA-binding molecule DRAQ5 were used to prepare anthraquinone-based compounds, which were assessed for their potential to interrogate cytochrome P450 (CYP) functional activity and localisation. Anthraquinones 124-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 227-230 29436156-0 2018 Cytochrome P450 Genetic Variation Associated with Tamoxifen Biotransformation in American Indian and Alaska Native People. Tamoxifen 50-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29436156-3 2018 We tested for associations between CYP diplotypes and plasma concentrations of tamoxifen and metabolites. Tamoxifen 79-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 29588191-1 2018 Few studies exist on cytochrome P450 (CYP450) metabolites of arachidonic acid (AA) pertaining to the pathophysiological events in pregnancy. Arachidonic Acid 61-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 29069987-1 2018 Cytochrome P450 enzymes are required for the synthesis of cholesterol and steroid hormones. Cholesterol 58-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29069987-1 2018 Cytochrome P450 enzymes are required for the synthesis of cholesterol and steroid hormones. Steroids 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29596975-0 2018 Role of cytochrome P450 enzymes in fimasartan metabolism in vitro. fimasartan 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 29596975-2 2018 The purpose of this study was to characterize enzymes involved in NADPH-dependent FMS metabolism using recombinant enzymes such as cytochrome P450 (CYP) and flavin-containing monooxygenase (FMO), as well as selective chemical inhibitors. NADP 66-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 29596975-2 2018 The purpose of this study was to characterize enzymes involved in NADPH-dependent FMS metabolism using recombinant enzymes such as cytochrome P450 (CYP) and flavin-containing monooxygenase (FMO), as well as selective chemical inhibitors. NADP 66-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 29588191-1 2018 Few studies exist on cytochrome P450 (CYP450) metabolites of arachidonic acid (AA) pertaining to the pathophysiological events in pregnancy. Arachidonic Acid 61-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-44 29698447-3 2018 It is now recognized that ARA is metabolized to a number of bioactive oxygenated lipids (oxylipins) by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP450) enzymes. Arachidonic Acid 26-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-164 29698447-7 2018 Individual CYP450- and LOX- but not COX-derived metabolites were higher with celecoxib than placebo (P<0.05) and differences were greater among non-aspirin users. Celecoxib 77-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-17 29698447-3 2018 It is now recognized that ARA is metabolized to a number of bioactive oxygenated lipids (oxylipins) by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP450) enzymes. Arachidonic Acid 26-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-172 29698447-9 2018 20-HETE, a prohypertensive androgen-sensitive CYP450 metabolite was higher with celecoxib absent aspirin and was positively associated with SBP in men (P = 0.040) but not women. Celecoxib 80-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-52 29659506-0 2018 In Vitro Inhibitory Effects of Synthetic Cannabinoid EAM-2201 on Cytochrome P450 and UDP-Glucuronosyltransferase Enzyme Activities in Human Liver Microsomes. Cannabinoids 41-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 29698447-9 2018 20-HETE, a prohypertensive androgen-sensitive CYP450 metabolite was higher with celecoxib absent aspirin and was positively associated with SBP in men (P = 0.040) but not women. Aspirin 97-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-52 29659506-0 2018 In Vitro Inhibitory Effects of Synthetic Cannabinoid EAM-2201 on Cytochrome P450 and UDP-Glucuronosyltransferase Enzyme Activities in Human Liver Microsomes. (4-ethyl-1-naphthalenyl)(1-(5-fluoropentyl)-1H-indol-3-yl)methanone 53-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 30181852-0 2018 Drug interactions of meglitinide antidiabetics involving CYP enzymes and OATP1B1 transporter. meglitinide 21-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 30181852-3 2018 Meglitinides are the substrates of cytochrome P450 (CYP) enzymes and organic anion transporting polypeptide 1B1 (OATP1B1 transporter) and the coadministration of the drugs affecting them will result in pharmacokinetic drug interactions. meglitinide 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 30181852-3 2018 Meglitinides are the substrates of cytochrome P450 (CYP) enzymes and organic anion transporting polypeptide 1B1 (OATP1B1 transporter) and the coadministration of the drugs affecting them will result in pharmacokinetic drug interactions. meglitinide 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 30181852-4 2018 This article focuses on the drug interactions of meglitinides involving CYP enzymes and OATP1B1 transporter. meglitinide 49-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 29143479-1 2018 Rifampin (RIF) is a bactericidal antibiotic drug and potent inducer of hepatic and intestinal cytochrome P-450 (CYP-450) enzyme systems. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 29670361-9 2018 Besides, the Kyoto Encyclopedia of Genes and Genomes pathways including chemical carcinogenesis, drug metabolism-cytochrome P450, tryptophan metabolism, and retinol metabolism were involved. Tryptophan 130-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 29670361-9 2018 Besides, the Kyoto Encyclopedia of Genes and Genomes pathways including chemical carcinogenesis, drug metabolism-cytochrome P450, tryptophan metabolism, and retinol metabolism were involved. Vitamin A 157-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 29143479-1 2018 Rifampin (RIF) is a bactericidal antibiotic drug and potent inducer of hepatic and intestinal cytochrome P-450 (CYP-450) enzyme systems. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-119 29143479-1 2018 Rifampin (RIF) is a bactericidal antibiotic drug and potent inducer of hepatic and intestinal cytochrome P-450 (CYP-450) enzyme systems. Rifampin 10-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 29143479-1 2018 Rifampin (RIF) is a bactericidal antibiotic drug and potent inducer of hepatic and intestinal cytochrome P-450 (CYP-450) enzyme systems. Rifampin 10-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-119 29105855-1 2018 AIM: The aim of this study was to study potential cytochrome P450 (CYP) induction by dicloxacillin. Dicloxacillin 85-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 28436281-2 2018 Roles of human cytochrome P450 (P450) 3A4 in oxidation of an antihistaminic drug terfenadine have been previously investigated in association with terfenadine-ketoconazole interaction. Terfenadine 81-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-41 28436281-2 2018 Roles of human cytochrome P450 (P450) 3A4 in oxidation of an antihistaminic drug terfenadine have been previously investigated in association with terfenadine-ketoconazole interaction. Terfenadine 147-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-41 28436281-2 2018 Roles of human cytochrome P450 (P450) 3A4 in oxidation of an antihistaminic drug terfenadine have been previously investigated in association with terfenadine-ketoconazole interaction. Ketoconazole 159-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-41 29277574-0 2018 Human cytochrome P450 kinetic studies on six N-2-methoxybenzyl (NBOMe)-derived new psychoactive substances using the substrate depletion approach. n-2-methoxybenzyl (nbome) 45-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 29277574-3 2018 Therefore, the aims of the present study were to determine the Km and Vmax values for CYP isoforms using the substrate depletion approach for the six N-2-methoxybenzyl (NBOMe)-derived NPS 25B-NBOMe, 25C-NBOMe, 25I-NBOMe, 3,4-DMA-NBOMe, 4-EA-NBOMe, and 4-MMA-NBOMe. 2-methoxybenzyl 152-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 29636974-4 2018 However, it is important to be aware that plasma levels of DOACs are affected by drugs that alter the cell efflux transporter P-glycoprotein and/or cytochrome P450. doacs 59-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-163 29105855-1 2018 AIM: The aim of this study was to study potential cytochrome P450 (CYP) induction by dicloxacillin. Dicloxacillin 85-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 29105855-8 2018 Investigations in primary hepatocytes showed a statistically significant dose-dependent increase in CYP expression and activity by dicloxacillin, caused by activation of the pregnane X receptor. Dicloxacillin 131-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 29255971-0 2018 Effects of a Fixed-Dose Co-Formulation of Daclatasvir, Asunaprevir, and Beclabuvir on the Pharmacokinetics of a Cocktail of Cytochrome P450 and Drug Transporter Substrates in Healthy Subjects. daclatasvir 42-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 29255971-0 2018 Effects of a Fixed-Dose Co-Formulation of Daclatasvir, Asunaprevir, and Beclabuvir on the Pharmacokinetics of a Cocktail of Cytochrome P450 and Drug Transporter Substrates in Healthy Subjects. asunaprevir 55-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 29255971-0 2018 Effects of a Fixed-Dose Co-Formulation of Daclatasvir, Asunaprevir, and Beclabuvir on the Pharmacokinetics of a Cocktail of Cytochrome P450 and Drug Transporter Substrates in Healthy Subjects. 8-cyclohexyl-N-((dimethylamino)sulfonyl)-1,1a,2,12b-tetrahydro-11-methoxy-1a-((3-methyl-3,8-diazabicyclo(3.2.1)oct-8-yl)carbonyl)cycloprop(d)indolo(2,1-a)(2)benzazepine-5-carboxamide 72-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 29080699-2 2018 This review covers recent findings indicating that a variety of these beneficial effects are mediated by "omega-3 epoxyeicosanoids", a class of novel n-3 LC-PUFA-derived lipid mediators, which are generated via the cytochrome P450 (CYP) epoxygenase pathway. omega-3 epoxyeicosanoids 106-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-230 29316516-4 2018 We also characterized fetal cytochrome P450 (CYP) mRNA expression and its associations with PBDE exposures. Halogenated Diphenyl Ethers 92-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 29316516-4 2018 We also characterized fetal cytochrome P450 (CYP) mRNA expression and its associations with PBDE exposures. Halogenated Diphenyl Ethers 92-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 29316516-14 2018 CONCLUSION: Our findings suggest that under normal conditions of mid-gestation, the human fetus is directly exposed to concentrations of PBDEs that may be higher than previously estimated based on maternal serum and that these exposures are associated with the expression of mRNAs coding for CYP enzymes. Halogenated Diphenyl Ethers 137-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 292-295 29080699-2 2018 This review covers recent findings indicating that a variety of these beneficial effects are mediated by "omega-3 epoxyeicosanoids", a class of novel n-3 LC-PUFA-derived lipid mediators, which are generated via the cytochrome P450 (CYP) epoxygenase pathway. omega-3 epoxyeicosanoids 106-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-235 29080699-3 2018 CYP enzymes, previously identified as arachidonic acid (20:4n-6; AA) epoxygenases, accept eicosapentaenoic acid (20:5n-3; EPA) and docosahexaenoic acid (22:6n-3; DHA), the major fish oil n-3 LC-PUFAs, as efficient alternative substrates. Arachidonic Acid 38-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29080699-3 2018 CYP enzymes, previously identified as arachidonic acid (20:4n-6; AA) epoxygenases, accept eicosapentaenoic acid (20:5n-3; EPA) and docosahexaenoic acid (22:6n-3; DHA), the major fish oil n-3 LC-PUFAs, as efficient alternative substrates. Eicosapentaenoic Acid 90-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29080699-3 2018 CYP enzymes, previously identified as arachidonic acid (20:4n-6; AA) epoxygenases, accept eicosapentaenoic acid (20:5n-3; EPA) and docosahexaenoic acid (22:6n-3; DHA), the major fish oil n-3 LC-PUFAs, as efficient alternative substrates. Docosahexaenoic Acids 131-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29080699-3 2018 CYP enzymes, previously identified as arachidonic acid (20:4n-6; AA) epoxygenases, accept eicosapentaenoic acid (20:5n-3; EPA) and docosahexaenoic acid (22:6n-3; DHA), the major fish oil n-3 LC-PUFAs, as efficient alternative substrates. dehydroacetic acid 162-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29080699-3 2018 CYP enzymes, previously identified as arachidonic acid (20:4n-6; AA) epoxygenases, accept eicosapentaenoic acid (20:5n-3; EPA) and docosahexaenoic acid (22:6n-3; DHA), the major fish oil n-3 LC-PUFAs, as efficient alternative substrates. Fatty Acids, Unsaturated 194-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 29080699-4 2018 In humans and rodents, dietary EPA/DHA supplementation causes a profound shift of the endogenous CYP-eicosanoid profile from AA- to EPA- and DHA-derived metabolites, increasing, in particular, the plasma and tissue levels of 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP). dehydroacetic acid 35-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 29080699-4 2018 In humans and rodents, dietary EPA/DHA supplementation causes a profound shift of the endogenous CYP-eicosanoid profile from AA- to EPA- and DHA-derived metabolites, increasing, in particular, the plasma and tissue levels of 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP). Eicosanoids 101-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 29080699-4 2018 In humans and rodents, dietary EPA/DHA supplementation causes a profound shift of the endogenous CYP-eicosanoid profile from AA- to EPA- and DHA-derived metabolites, increasing, in particular, the plasma and tissue levels of 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP). dehydroacetic acid 141-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 29080699-4 2018 In humans and rodents, dietary EPA/DHA supplementation causes a profound shift of the endogenous CYP-eicosanoid profile from AA- to EPA- and DHA-derived metabolites, increasing, in particular, the plasma and tissue levels of 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP). 17,18-epoxy-5,8,11,14-eicosatetraenoic acid 225-257 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 29080699-4 2018 In humans and rodents, dietary EPA/DHA supplementation causes a profound shift of the endogenous CYP-eicosanoid profile from AA- to EPA- and DHA-derived metabolites, increasing, in particular, the plasma and tissue levels of 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP). 17(18)-EpETE 259-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 29080699-4 2018 In humans and rodents, dietary EPA/DHA supplementation causes a profound shift of the endogenous CYP-eicosanoid profile from AA- to EPA- and DHA-derived metabolites, increasing, in particular, the plasma and tissue levels of 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP). 19,20-epoxydocosapentaenoic acid 274-306 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 29080699-4 2018 In humans and rodents, dietary EPA/DHA supplementation causes a profound shift of the endogenous CYP-eicosanoid profile from AA- to EPA- and DHA-derived metabolites, increasing, in particular, the plasma and tissue levels of 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP). 19(20)-EpDPE 308-317 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 29479969-0 2018 Detection of Cytochrome P450 Polymorphisms in BreastCancer Patients May Impact on Tamoxifen Therapy Background: Breast cancer is the most common cancer among women worldwide. Tamoxifen 82-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 29479969-2 2018 TAM is metabolized by cytochrome P450 (CYP450) enzymes,including CYP2D6, CYP3A5 and CYP2C19, whose genetic variations may have clinicopathological importance.However, reports on the association of various P450 polymorphisms with certain cancers are contradictory. Tamoxifen 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 29479969-2 2018 TAM is metabolized by cytochrome P450 (CYP450) enzymes,including CYP2D6, CYP3A5 and CYP2C19, whose genetic variations may have clinicopathological importance.However, reports on the association of various P450 polymorphisms with certain cancers are contradictory. Tamoxifen 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-45 28668640-1 2018 The cytochrome P450 monooxygenases (P450s) are thiolate heme proteins that can, often under physiological conditions, catalyze many distinct oxidative transformations on a wide variety of molecules, including relatively simple alkanes or fatty acids, as well as more complex compounds such as steroids and exogenous pollutants. thiolate 47-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 29266939-1 2018 Cytochrome P450 (CYP) monooxygenases catalyze the oxidation of chemically inert carbon-hydrogen bonds in diverse endogenous and exogenous organic compounds by atmospheric oxygen. Carbon 80-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29266939-1 2018 Cytochrome P450 (CYP) monooxygenases catalyze the oxidation of chemically inert carbon-hydrogen bonds in diverse endogenous and exogenous organic compounds by atmospheric oxygen. Carbon 80-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 29266939-1 2018 Cytochrome P450 (CYP) monooxygenases catalyze the oxidation of chemically inert carbon-hydrogen bonds in diverse endogenous and exogenous organic compounds by atmospheric oxygen. Hydrogen 87-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29266939-1 2018 Cytochrome P450 (CYP) monooxygenases catalyze the oxidation of chemically inert carbon-hydrogen bonds in diverse endogenous and exogenous organic compounds by atmospheric oxygen. Hydrogen 87-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 29266939-1 2018 Cytochrome P450 (CYP) monooxygenases catalyze the oxidation of chemically inert carbon-hydrogen bonds in diverse endogenous and exogenous organic compounds by atmospheric oxygen. Oxygen 26-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29266939-1 2018 Cytochrome P450 (CYP) monooxygenases catalyze the oxidation of chemically inert carbon-hydrogen bonds in diverse endogenous and exogenous organic compounds by atmospheric oxygen. Oxygen 26-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 29527583-1 2018 The cytochrome P450 (CYP) enzymes are a diverse group of heme monooxygenases that, through the course of their reaction cycle, contribute to cellular reactive oxygen species (ROS). Reactive Oxygen Species 150-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 29527583-1 2018 The cytochrome P450 (CYP) enzymes are a diverse group of heme monooxygenases that, through the course of their reaction cycle, contribute to cellular reactive oxygen species (ROS). Reactive Oxygen Species 150-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 29527583-1 2018 The cytochrome P450 (CYP) enzymes are a diverse group of heme monooxygenases that, through the course of their reaction cycle, contribute to cellular reactive oxygen species (ROS). Reactive Oxygen Species 175-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 29527583-1 2018 The cytochrome P450 (CYP) enzymes are a diverse group of heme monooxygenases that, through the course of their reaction cycle, contribute to cellular reactive oxygen species (ROS). Reactive Oxygen Species 175-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 29527583-3 2018 However, during the course of the CYP catalytic cycle, ROS can be generated through uncoupling of the enzymatic cycle. Reactive Oxygen Species 55-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 28473297-0 2018 NADH reduction of nitroaromatics as a probe for residual ferric form high-spin in a cytochrome P450. NAD 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 28473297-0 2018 NADH reduction of nitroaromatics as a probe for residual ferric form high-spin in a cytochrome P450. nitroaromatics 18-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 28668640-1 2018 The cytochrome P450 monooxygenases (P450s) are thiolate heme proteins that can, often under physiological conditions, catalyze many distinct oxidative transformations on a wide variety of molecules, including relatively simple alkanes or fatty acids, as well as more complex compounds such as steroids and exogenous pollutants. Alkanes 227-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 28473297-0 2018 NADH reduction of nitroaromatics as a probe for residual ferric form high-spin in a cytochrome P450. Ferric enterobactin ion 57-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 28668640-1 2018 The cytochrome P450 monooxygenases (P450s) are thiolate heme proteins that can, often under physiological conditions, catalyze many distinct oxidative transformations on a wide variety of molecules, including relatively simple alkanes or fatty acids, as well as more complex compounds such as steroids and exogenous pollutants. Fatty Acids 238-249 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 28473297-1 2018 The existence of a substrate-sensitive equilibrium between high spin (S=5/2) and low spin (S=1/2) ferric iron is a well-established phenomenon in the cytochrome P450 (CYP) superfamily, although its origins are still a subject of discussion. ferric sulfate 98-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-165 28473297-1 2018 The existence of a substrate-sensitive equilibrium between high spin (S=5/2) and low spin (S=1/2) ferric iron is a well-established phenomenon in the cytochrome P450 (CYP) superfamily, although its origins are still a subject of discussion. ferric sulfate 98-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-170 28668640-1 2018 The cytochrome P450 monooxygenases (P450s) are thiolate heme proteins that can, often under physiological conditions, catalyze many distinct oxidative transformations on a wide variety of molecules, including relatively simple alkanes or fatty acids, as well as more complex compounds such as steroids and exogenous pollutants. Steroids 293-301 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 28473297-7 2018 These results suggest that reduction of nitroaromatics may provide a useful probe of residual high spin states in the CYP superfamily. nitroaromatics 40-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 28865153-1 2018 AIMS: This phase 1, open-label, crossover study sought to evaluate drug-drug interactions between tivantinib and cytochrome P450 (CYP) substrates and tivantinib and P-glycoprotein. ARQ 197 98-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 29984664-0 2018 Genistein Affects Expression of Cytochrome P450 (CYP450) Genes in Hepatocellular Carcinoma (HEPG2/C3A) Cell Line. Genistein 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 27637911-2 2018 The aim of this study was to investigate the association between cytochrome P450 ( CYP) genetic variants and clinical adverse outcomes of concomitant use of PPIs and clopidogrel by patients. Clopidogrel 166-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 27637911-2 2018 The aim of this study was to investigate the association between cytochrome P450 ( CYP) genetic variants and clinical adverse outcomes of concomitant use of PPIs and clopidogrel by patients. Clopidogrel 166-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 28118749-0 2018 Warfarin Interaction With Hepatic Cytochrome P-450 Enzyme-Inducing Anticonvulsants. Warfarin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 28118749-1 2018 Initiation of cytochrome P-450 (CYP)-inducing anticonvulsant medications during warfarin therapy may decrease anticoagulant effect and necessitate frequent warfarin dose adjustments to maintain therapeutic response measured by the international normalized ratio (INR). Warfarin 80-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 28118749-1 2018 Initiation of cytochrome P-450 (CYP)-inducing anticonvulsant medications during warfarin therapy may decrease anticoagulant effect and necessitate frequent warfarin dose adjustments to maintain therapeutic response measured by the international normalized ratio (INR). Warfarin 80-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 28118749-1 2018 Initiation of cytochrome P-450 (CYP)-inducing anticonvulsant medications during warfarin therapy may decrease anticoagulant effect and necessitate frequent warfarin dose adjustments to maintain therapeutic response measured by the international normalized ratio (INR). Warfarin 156-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 28118749-1 2018 Initiation of cytochrome P-450 (CYP)-inducing anticonvulsant medications during warfarin therapy may decrease anticoagulant effect and necessitate frequent warfarin dose adjustments to maintain therapeutic response measured by the international normalized ratio (INR). Warfarin 156-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 27875967-1 2018 Cytochrome P450 enzymes are a structurally conserved but functionally diverse group of heme-containing mixed function oxidases found across both prokaryotic and eukaryotic forms of the microbial world. Heme 87-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Omeprazole 146-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Omeprazole 158-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Phenobarbital 175-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Phenobarbital 190-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Rifampin 210-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 29357810-5 2018 mRNA expression levels and enzyme activities of CYP1A2, CYP2B6, and CYP3A in HepaRG cells treated with prototypical inducers of each CYP isoform [omeprazole (OME) for CYP1A2, phenobarbital (PB) for CYP2B6, and rifampicin (RIF) for CYP3A] were evaluated. Rifampin 222-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 29984664-2 2018 Of particular interest regarding cancer preventive properties of flavonoids is their interaction with cytochrome P450 enzymes (CYPs). Flavonoids 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-117 29984664-4 2018 OBJECTIVE: The aim of this study was to investigate the effects of genistein on cytochrome P450 (CYP) gene expression levels in human hepatocellular carcinoma (HepG2/C3A) and colon adenocarcinoma (HT29) cells. Genistein 67-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 29984664-4 2018 OBJECTIVE: The aim of this study was to investigate the effects of genistein on cytochrome P450 (CYP) gene expression levels in human hepatocellular carcinoma (HepG2/C3A) and colon adenocarcinoma (HT29) cells. Genistein 67-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 29491651-0 2018 Studying the Inhibitory Effect of Quercetin and Thymoquinone on Human Cytochrome P450 Enzyme Activities. Quercetin 34-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 29555536-8 2018 The results demonstrate that the xenobiotic metabolism conferred by transfection of CYP-encoding mRNAs shifts the dose-response relationship for some of the tested chemicals such as aflatoxin B1 (bioactivation) and fenazaquin (detoxification). Aflatoxin B1 182-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 29555536-8 2018 The results demonstrate that the xenobiotic metabolism conferred by transfection of CYP-encoding mRNAs shifts the dose-response relationship for some of the tested chemicals such as aflatoxin B1 (bioactivation) and fenazaquin (detoxification). fenazaquin 215-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 29393278-0 2018 Effect of Oridonin on Cytochrome P450 Expression and Activities in HepaRG Cell. oridonin 10-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 29618712-3 2018 In this study, however, after 3-4 weeks of culture according to the manufacturer"s instructions, human cytochrome P450 (P450) 2C9- and 2C19-dependent diclofenac 4"-hydroxylation and omeprazole 5-hydroxylation activities of the iPS-derived hepatocytes had significantly increased above the activities at 1 week and had reached levels similar to those in HepaRG cells, a human hepatocyte-like cell line. diclofenac 4" 150-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-129 29618712-3 2018 In this study, however, after 3-4 weeks of culture according to the manufacturer"s instructions, human cytochrome P450 (P450) 2C9- and 2C19-dependent diclofenac 4"-hydroxylation and omeprazole 5-hydroxylation activities of the iPS-derived hepatocytes had significantly increased above the activities at 1 week and had reached levels similar to those in HepaRG cells, a human hepatocyte-like cell line. IPS 227-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-129 29491651-0 2018 Studying the Inhibitory Effect of Quercetin and Thymoquinone on Human Cytochrome P450 Enzyme Activities. thymoquinone 48-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 29503429-4 2018 Our previous studies revealed that sesamin was sequentially metabolized by cytochrome P450 (CYP) and UDP-glucuronosyltransferase or sulfotransferase. sesamin 35-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 29503429-4 2018 Our previous studies revealed that sesamin was sequentially metabolized by cytochrome P450 (CYP) and UDP-glucuronosyltransferase or sulfotransferase. sesamin 35-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 29024391-1 2017 A cytochrome P450 was engineered to selectively incorporate Ir(Me)-deuteroporphyrin IX (Ir(Me)-DPIX), in lieu of heme, in bacterial cells. deuteroporphyrin-IX 67-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-17 29024391-1 2017 A cytochrome P450 was engineered to selectively incorporate Ir(Me)-deuteroporphyrin IX (Ir(Me)-DPIX), in lieu of heme, in bacterial cells. dpix 95-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-17 29024391-1 2017 A cytochrome P450 was engineered to selectively incorporate Ir(Me)-deuteroporphyrin IX (Ir(Me)-DPIX), in lieu of heme, in bacterial cells. Heme 113-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-17 29075787-9 2017 We suggest that intracellularly generated salicylic acid metabolites through CYP450 enzymes within the colonic epithelial cells, or the salicylic acid metabolites generated by gut microflora may significantly contribute to the preferential chemopreventive effect of aspirin against CRC through inhibition of CDKs. Salicylic Acid 42-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-83 29131622-1 2017 Cytochrome P450 enzymes were recently engineered to catalyze the C-H amination reaction of aryl sulfonyl azides with excellent regio- and stereoselectivity (Arnold and co-workers J. aryl sulfonyl azides 91-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28702763-6 2017 Azoles are substrates and inhibitors of cytochrome P450 (CYP) isoenzymes and are therefore involved in numerous drug-drug interactions. Azoles 0-6 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 28702763-6 2017 Azoles are substrates and inhibitors of cytochrome P450 (CYP) isoenzymes and are therefore involved in numerous drug-drug interactions. Azoles 0-6 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 29075787-3 2017 Our goal here was to investigate the ability of salicylic acid metabolites, known to be generated through cytochrome P450 (CYP450) enzymes, and its derivatives as cyclin dependent kinase (CDK) inhibitors to gain new insights into aspirin"s chemopreventive actions. Salicylic Acid 48-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 29075787-3 2017 Our goal here was to investigate the ability of salicylic acid metabolites, known to be generated through cytochrome P450 (CYP450) enzymes, and its derivatives as cyclin dependent kinase (CDK) inhibitors to gain new insights into aspirin"s chemopreventive actions. Salicylic Acid 48-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-129 28872689-5 2017 METHODS: Two flavonoids, luteolin and quercetin, were evaluated as potential inhibitors of eight human CYP isoforms, of six UDP-glucuronosyltransferase (UGT) isoforms and of APAP glucuronidation and sulfation. Flavonoids 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 29075787-3 2017 Our goal here was to investigate the ability of salicylic acid metabolites, known to be generated through cytochrome P450 (CYP450) enzymes, and its derivatives as cyclin dependent kinase (CDK) inhibitors to gain new insights into aspirin"s chemopreventive actions. Aspirin 230-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 29075787-3 2017 Our goal here was to investigate the ability of salicylic acid metabolites, known to be generated through cytochrome P450 (CYP450) enzymes, and its derivatives as cyclin dependent kinase (CDK) inhibitors to gain new insights into aspirin"s chemopreventive actions. Aspirin 230-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-129 28872689-5 2017 METHODS: Two flavonoids, luteolin and quercetin, were evaluated as potential inhibitors of eight human CYP isoforms, of six UDP-glucuronosyltransferase (UGT) isoforms and of APAP glucuronidation and sulfation. Luteolin 25-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 28872689-5 2017 METHODS: Two flavonoids, luteolin and quercetin, were evaluated as potential inhibitors of eight human CYP isoforms, of six UDP-glucuronosyltransferase (UGT) isoforms and of APAP glucuronidation and sulfation. Quercetin 38-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 28872689-7 2017 KEY FINDINGS: Luteolin and quercetin inhibited human CYP isoforms to varying degrees, with greatest potency towards CYP1A2 and CYP2C8. Quercetin 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 28872689-11 2017 CONCLUSIONS: Inhibition of human CYP activity by luteolin and quercetin occurred with IC50 values exceeding customary in-vivo human exposure with tolerable supplemental doses of these compounds. Quercetin 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 27841077-3 2017 The purpose of this study was to evaluate in vitro the potential to inhibit and induce cytochrome P450 (CYP) isoforms for alectinib and its major metabolite M4. alectinib 122-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 28614988-3 2017 However, whether DHM affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. dihydromyricetin 17-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 28614988-3 2017 However, whether DHM affects the activity of human liver cytochrome P450 (CYP) enzymes remains unclear. dihydromyricetin 17-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 28614988-4 2017 MATERIALS AND METHODS: The inhibitory effects of DHM on eight human liver CYP isoforms (i.e., 1A2, 3A4, 2A6, 2E1, 2D6, 2C9, 2C19 and 2C8) were investigated in vitro using human liver microsomes (HLMs). dihydromyricetin 49-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 28614988-5 2017 RESULTS: The results showed that DHM could inhibit the activity of CYP3A4, CYP2E1 and CYP2D6, with IC50 values of 14.75, 25.74 and 22.69 muM, respectively, but that other CYP isoforms were not affected. dihydromyricetin 33-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 28614988-8 2017 DISCUSSION AND CONCLUSION: The in vitro studies of DHM with CYP isoforms indicate that DHM has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP3A4, CYP2E1 and CYP2D6. dihydromyricetin 51-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 28614988-8 2017 DISCUSSION AND CONCLUSION: The in vitro studies of DHM with CYP isoforms indicate that DHM has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP3A4, CYP2E1 and CYP2D6. dihydromyricetin 87-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 28614988-0 2017 In vitro inhibitory effects of dihydromyricetin on human liver cytochrome P450 enzymes. dihydromyricetin 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 29143058-1 2017 Heme"s spin-multiplicity is key in determining the enzymatic function of cytochrome P450 (cytP450). Heme 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 29143058-1 2017 Heme"s spin-multiplicity is key in determining the enzymatic function of cytochrome P450 (cytP450). Heme 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-97 28947469-1 2017 Nevirapine is metabolized by several hepatic cytochrome P450 (CYP) isoforms to generate four primary hydroxylated metabolites: 2-hydroxynevirapine, 3-hydroxynevirapine, 8-hydroxynevirapine, and 12-hydroxynevirapine. Nevirapine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 28947469-1 2017 Nevirapine is metabolized by several hepatic cytochrome P450 (CYP) isoforms to generate four primary hydroxylated metabolites: 2-hydroxynevirapine, 3-hydroxynevirapine, 8-hydroxynevirapine, and 12-hydroxynevirapine. Nevirapine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 28947469-1 2017 Nevirapine is metabolized by several hepatic cytochrome P450 (CYP) isoforms to generate four primary hydroxylated metabolites: 2-hydroxynevirapine, 3-hydroxynevirapine, 8-hydroxynevirapine, and 12-hydroxynevirapine. 2-Hydroxy Nevirapine 127-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 28947469-1 2017 Nevirapine is metabolized by several hepatic cytochrome P450 (CYP) isoforms to generate four primary hydroxylated metabolites: 2-hydroxynevirapine, 3-hydroxynevirapine, 8-hydroxynevirapine, and 12-hydroxynevirapine. 2-Hydroxy Nevirapine 127-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 27841077-3 2017 The purpose of this study was to evaluate in vitro the potential to inhibit and induce cytochrome P450 (CYP) isoforms for alectinib and its major metabolite M4. alectinib 122-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-107 27841077-8 2017 Out of the seven CYP isoforms in HLM, alectinib and M4 showed time-dependent inhibition (TDI) of only CYP3A4, which suggests low TDI potential due to low inactivation efficiency. alectinib 38-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 28947469-1 2017 Nevirapine is metabolized by several hepatic cytochrome P450 (CYP) isoforms to generate four primary hydroxylated metabolites: 2-hydroxynevirapine, 3-hydroxynevirapine, 8-hydroxynevirapine, and 12-hydroxynevirapine. 8-Hydroxynevirapine 169-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 28947469-1 2017 Nevirapine is metabolized by several hepatic cytochrome P450 (CYP) isoforms to generate four primary hydroxylated metabolites: 2-hydroxynevirapine, 3-hydroxynevirapine, 8-hydroxynevirapine, and 12-hydroxynevirapine. 8-Hydroxynevirapine 169-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 27841077-12 2017 In summary, the risk of alectinib causing drug-drug interactions with coadministered drugs is expected to be low due to the weak potential of CYP inhibition and induction estimated in the preclinical studies. alectinib 24-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-145 28947469-1 2017 Nevirapine is metabolized by several hepatic cytochrome P450 (CYP) isoforms to generate four primary hydroxylated metabolites: 2-hydroxynevirapine, 3-hydroxynevirapine, 8-hydroxynevirapine, and 12-hydroxynevirapine. 12-Hydroxynevirapine 194-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 28947469-1 2017 Nevirapine is metabolized by several hepatic cytochrome P450 (CYP) isoforms to generate four primary hydroxylated metabolites: 2-hydroxynevirapine, 3-hydroxynevirapine, 8-hydroxynevirapine, and 12-hydroxynevirapine. 12-Hydroxynevirapine 194-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 29170672-0 2017 Triterpene Structural Diversification by Plant Cytochrome P450 Enzymes. Triterpenes 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 28431455-6 2017 In addition, the specific cytochrome P450 (CYP) enzymes involved in the metabolism of cnidilin were identified using chemical inhibition and CYP recombinant enzymes. cnidilin 86-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 28431455-6 2017 In addition, the specific cytochrome P450 (CYP) enzymes involved in the metabolism of cnidilin were identified using chemical inhibition and CYP recombinant enzymes. cnidilin 86-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 28431455-6 2017 In addition, the specific cytochrome P450 (CYP) enzymes involved in the metabolism of cnidilin were identified using chemical inhibition and CYP recombinant enzymes. cnidilin 86-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-144 29022838-4 2017 Areas covered: This manuscript reviews the pharmacogenomics (i.e. the influence of genetics on drug disposition) of triazole antifungal agents related to their CYP-mediated metabolism and summarizes their implications on triazole efficacy, safety, and tolerability. Triazoles 116-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-163 28856745-5 2017 RESULTS: A total of 10 publications were identified in which a CYP450 enzyme inducer was utilized intentionally to enhance CNI clearance in the setting of supratherapeutic concentrations; 7 case reports describe the use of phenytoin and 3 case reports describe the use of phenobarbital. Phenytoin 223-232 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-69 28856745-5 2017 RESULTS: A total of 10 publications were identified in which a CYP450 enzyme inducer was utilized intentionally to enhance CNI clearance in the setting of supratherapeutic concentrations; 7 case reports describe the use of phenytoin and 3 case reports describe the use of phenobarbital. Phenobarbital 272-285 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-69 29022838-4 2017 Areas covered: This manuscript reviews the pharmacogenomics (i.e. the influence of genetics on drug disposition) of triazole antifungal agents related to their CYP-mediated metabolism and summarizes their implications on triazole efficacy, safety, and tolerability. Triazoles 221-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-163 29053951-0 2017 Heme Binding Biguanides Target Cytochrome P450-Dependent Cancer Cell Mitochondria. Heme 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 28527918-0 2017 Cytochrome P450 eicosanoids in cerebrovascular function and disease. Eicosanoids 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28527918-1 2017 Cytochrome P450 eicosanoids play important roles in brain function and disease through their complementary actions on cell-cell communications within the neurovascular unit (NVU) and mechanisms of brain injury. Eicosanoids 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28527918-2 2017 Epoxy- and hydroxyeicosanoids, respectively formed by cytochrome P450 epoxygenases and omega-hydroxylases, play opposing roles in cerebrovascular function and in pathological processes underlying neural injury, including ischemia, neuroinflammation and oxidative injury. epoxy- and hydroxyeicosanoids 0-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 28756170-0 2017 Confirmation of metabolites of the neuroleptic drug prothipendyl using human liver microsomes, specific CYP enzymes and authentic forensic samples-Benefit for routine drug testing. prothipendyl 52-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-107 28756170-1 2017 Metabolism of the tricyclic azaphenothiazine neuroleptic drug prothipendyl was investigated with in vitro studies using human liver microsomes but also specific isoforms of cytochrome P450 (CYP) enzymes. azaphenothiazine 28-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-188 28756170-1 2017 Metabolism of the tricyclic azaphenothiazine neuroleptic drug prothipendyl was investigated with in vitro studies using human liver microsomes but also specific isoforms of cytochrome P450 (CYP) enzymes. azaphenothiazine 28-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-193 28756170-1 2017 Metabolism of the tricyclic azaphenothiazine neuroleptic drug prothipendyl was investigated with in vitro studies using human liver microsomes but also specific isoforms of cytochrome P450 (CYP) enzymes. prothipendyl 62-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-188 28756170-1 2017 Metabolism of the tricyclic azaphenothiazine neuroleptic drug prothipendyl was investigated with in vitro studies using human liver microsomes but also specific isoforms of cytochrome P450 (CYP) enzymes. prothipendyl 62-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-193 28756170-5 2017 N-demethyl-prothipendyl was predominantly formed by isoforms CYP2C19 and CYP1A2, while particularly the CYP isoenzyme 3A4 was responsible for the formation of prothipendyl sulfoxide. n-demethyl-prothipendyl 0-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 29065189-6 2017 Furthermore, preincubation of TC-HepG2 cells with CYP inhibitors known as time-dependent inhibitors (TDI) prior to the addition of CYP-specific substrates determined that CYP inhibition was enhanced in the TDI group than in the non-TDI group. Toluene 2,4-Diisocyanate 101-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 29065189-6 2017 Furthermore, preincubation of TC-HepG2 cells with CYP inhibitors known as time-dependent inhibitors (TDI) prior to the addition of CYP-specific substrates determined that CYP inhibition was enhanced in the TDI group than in the non-TDI group. Toluene 2,4-Diisocyanate 206-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 29065189-6 2017 Furthermore, preincubation of TC-HepG2 cells with CYP inhibitors known as time-dependent inhibitors (TDI) prior to the addition of CYP-specific substrates determined that CYP inhibition was enhanced in the TDI group than in the non-TDI group. Toluene 2,4-Diisocyanate 206-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 29065189-6 2017 Furthermore, preincubation of TC-HepG2 cells with CYP inhibitors known as time-dependent inhibitors (TDI) prior to the addition of CYP-specific substrates determined that CYP inhibition was enhanced in the TDI group than in the non-TDI group. Toluene 2,4-Diisocyanate 206-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 28919040-0 2017 Heme Binding Biguanides Target Cytochrome P450-Dependent Cancer Cell Mitochondria. Heme 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 28919040-0 2017 Heme Binding Biguanides Target Cytochrome P450-Dependent Cancer Cell Mitochondria. Biguanides 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 29053951-0 2017 Heme Binding Biguanides Target Cytochrome P450-Dependent Cancer Cell Mitochondria. Biguanides 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 28960974-5 2017 We pinpointed numerous reactive cysteines as the targets of RMs of diclofenac, including the active (heme-binding) sites on several key CYP450 isoforms (1A2, 2E1 and 3A4 for human, 2C39 and 3A11 for mouse). Diclofenac 67-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-142 28960974-5 2017 We pinpointed numerous reactive cysteines as the targets of RMs of diclofenac, including the active (heme-binding) sites on several key CYP450 isoforms (1A2, 2E1 and 3A4 for human, 2C39 and 3A11 for mouse). Heme 101-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-142 28701518-0 2017 Effect of Cytochrome P450 Metabolites of Arachidonic Acid in Nephrology. Arachidonic Acid 41-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 28677033-1 2017 Due to intestinal cytochrome P450 (CYP450)-mediated metabolism and P-glycoprotein (P-gp) efflux, poor oral bioavailability hinders ginsenoside-Rh1 (Rh1) and ginsenoside-Rh2 (Rh2) from clinical application. ginsenoside Rh1 131-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 28677033-1 2017 Due to intestinal cytochrome P450 (CYP450)-mediated metabolism and P-glycoprotein (P-gp) efflux, poor oral bioavailability hinders ginsenoside-Rh1 (Rh1) and ginsenoside-Rh2 (Rh2) from clinical application. ginsenoside Rh1 131-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-41 28677033-1 2017 Due to intestinal cytochrome P450 (CYP450)-mediated metabolism and P-glycoprotein (P-gp) efflux, poor oral bioavailability hinders ginsenoside-Rh1 (Rh1) and ginsenoside-Rh2 (Rh2) from clinical application. 2,5-diaziridinyl-3-(hydroxymethyl)-6-methyl-1,4-benzoquinone 143-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 28677033-1 2017 Due to intestinal cytochrome P450 (CYP450)-mediated metabolism and P-glycoprotein (P-gp) efflux, poor oral bioavailability hinders ginsenoside-Rh1 (Rh1) and ginsenoside-Rh2 (Rh2) from clinical application. 2,5-diaziridinyl-3-(hydroxymethyl)-6-methyl-1,4-benzoquinone 143-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-41 28677033-7 2017 We confirmed that SME containing CYP450 and P-gp inhibitory excipient could distinctively improve the oral availabilities of Rh1 compared to free drug or SME containing P-gp inhibitory excipient. 8-O-acetyl shanzhiside methyl ester 18-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-39 28185143-1 2017 BACKGROUND AND OBJECTIVES: 1-Aminobenzotriazole, a known time-dependent inhibitor of cytochrome P450 (CYP) enzymes, and ketoconazole, a strong inhibitor of the human CYP3A4 isozyme, are used as standard probe inhibitors to characterize the CYP and/or non-CYP-mediated metabolism of xenobiotics. 1-aminobenzotriazole 27-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 28185143-1 2017 BACKGROUND AND OBJECTIVES: 1-Aminobenzotriazole, a known time-dependent inhibitor of cytochrome P450 (CYP) enzymes, and ketoconazole, a strong inhibitor of the human CYP3A4 isozyme, are used as standard probe inhibitors to characterize the CYP and/or non-CYP-mediated metabolism of xenobiotics. 1-aminobenzotriazole 27-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 28185143-1 2017 BACKGROUND AND OBJECTIVES: 1-Aminobenzotriazole, a known time-dependent inhibitor of cytochrome P450 (CYP) enzymes, and ketoconazole, a strong inhibitor of the human CYP3A4 isozyme, are used as standard probe inhibitors to characterize the CYP and/or non-CYP-mediated metabolism of xenobiotics. 1-aminobenzotriazole 27-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-169 28185143-1 2017 BACKGROUND AND OBJECTIVES: 1-Aminobenzotriazole, a known time-dependent inhibitor of cytochrome P450 (CYP) enzymes, and ketoconazole, a strong inhibitor of the human CYP3A4 isozyme, are used as standard probe inhibitors to characterize the CYP and/or non-CYP-mediated metabolism of xenobiotics. 1-aminobenzotriazole 27-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-169 28185143-1 2017 BACKGROUND AND OBJECTIVES: 1-Aminobenzotriazole, a known time-dependent inhibitor of cytochrome P450 (CYP) enzymes, and ketoconazole, a strong inhibitor of the human CYP3A4 isozyme, are used as standard probe inhibitors to characterize the CYP and/or non-CYP-mediated metabolism of xenobiotics. Ketoconazole 120-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 28185143-1 2017 BACKGROUND AND OBJECTIVES: 1-Aminobenzotriazole, a known time-dependent inhibitor of cytochrome P450 (CYP) enzymes, and ketoconazole, a strong inhibitor of the human CYP3A4 isozyme, are used as standard probe inhibitors to characterize the CYP and/or non-CYP-mediated metabolism of xenobiotics. Ketoconazole 120-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-169 28185143-1 2017 BACKGROUND AND OBJECTIVES: 1-Aminobenzotriazole, a known time-dependent inhibitor of cytochrome P450 (CYP) enzymes, and ketoconazole, a strong inhibitor of the human CYP3A4 isozyme, are used as standard probe inhibitors to characterize the CYP and/or non-CYP-mediated metabolism of xenobiotics. Ketoconazole 120-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-169 28767146-0 2017 A Randomized Trial of Effects of Alcohol on Cytochrome P450 Eicosanoids, Mediators of Inflammation Resolution, and Blood Pressure in Men. Alcohols 33-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 28767146-0 2017 A Randomized Trial of Effects of Alcohol on Cytochrome P450 Eicosanoids, Mediators of Inflammation Resolution, and Blood Pressure in Men. Eicosanoids 60-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 28767146-2 2017 We previously showed that chronic alcohol consumption increased blood pressure (BP), oxidative stress, and 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoconstrictor and pro-inflammatory eicosanoid synthesized by cytochrome P450 (CYP450) enzymes from arachidonic acid. Alcohols 34-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-230 28767146-2 2017 We previously showed that chronic alcohol consumption increased blood pressure (BP), oxidative stress, and 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoconstrictor and pro-inflammatory eicosanoid synthesized by cytochrome P450 (CYP450) enzymes from arachidonic acid. Alcohols 34-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-238 28767146-2 2017 We previously showed that chronic alcohol consumption increased blood pressure (BP), oxidative stress, and 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoconstrictor and pro-inflammatory eicosanoid synthesized by cytochrome P450 (CYP450) enzymes from arachidonic acid. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 107-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-230 28767146-2 2017 We previously showed that chronic alcohol consumption increased blood pressure (BP), oxidative stress, and 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoconstrictor and pro-inflammatory eicosanoid synthesized by cytochrome P450 (CYP450) enzymes from arachidonic acid. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 107-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-238 28767146-2 2017 We previously showed that chronic alcohol consumption increased blood pressure (BP), oxidative stress, and 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoconstrictor and pro-inflammatory eicosanoid synthesized by cytochrome P450 (CYP450) enzymes from arachidonic acid. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 140-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-230 28767146-2 2017 We previously showed that chronic alcohol consumption increased blood pressure (BP), oxidative stress, and 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoconstrictor and pro-inflammatory eicosanoid synthesized by cytochrome P450 (CYP450) enzymes from arachidonic acid. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 140-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-238 28767146-2 2017 We previously showed that chronic alcohol consumption increased blood pressure (BP), oxidative stress, and 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoconstrictor and pro-inflammatory eicosanoid synthesized by cytochrome P450 (CYP450) enzymes from arachidonic acid. Eicosanoids 189-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-230 28767146-2 2017 We previously showed that chronic alcohol consumption increased blood pressure (BP), oxidative stress, and 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoconstrictor and pro-inflammatory eicosanoid synthesized by cytochrome P450 (CYP450) enzymes from arachidonic acid. Eicosanoids 189-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-238 28767146-2 2017 We previously showed that chronic alcohol consumption increased blood pressure (BP), oxidative stress, and 20-hydroxyeicosatetraenoic acid (20-HETE), a vasoconstrictor and pro-inflammatory eicosanoid synthesized by cytochrome P450 (CYP450) enzymes from arachidonic acid. Arachidonic Acid 253-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-230 28767146-10 2017 These paradoxical findings require further studies to determine whether alcohol stimulates different CYP450 enzymes and whether the findings can be replicated in females. Alcohols 72-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 28701518-1 2017 Thirty-five years ago, a third pathway for the metabolism of arachidonic acid by cytochrome P450 enzymes emerged. Arachidonic Acid 61-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 28701518-4 2017 Studies in animal models suggest that changes in the production of cytochrome P450 eicosanoids alter BP. Eicosanoids 83-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 28701518-4 2017 Studies in animal models suggest that changes in the production of cytochrome P450 eicosanoids alter BP. Benzo(a)pyrene 101-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 28701518-8 2017 Our review summarizes the emerging evidence that cytochrome P450 eicosanoids have a role in the pathogenesis of hypertension, polycystic kidney disease, AKI, and CKD. Eicosanoids 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 29183231-1 2017 The cytochrome P450 (CYP) enzyme superfamily is involved in phase I metabolism which chemically modifies a variety of substrates via oxidative reactions to make them more water-soluble and easier to eliminate. Water 171-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 29183231-1 2017 The cytochrome P450 (CYP) enzyme superfamily is involved in phase I metabolism which chemically modifies a variety of substrates via oxidative reactions to make them more water-soluble and easier to eliminate. Water 171-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 28886714-9 2017 Observed changes in oxylipin levels can be connected to activation of cytochrome P450 (CYP) and 5-lipoxygenase (5-LOX) metabolic pathways in malaria infected individuals compared to controls, and related to increased levels of all linoleic acid oxylipins in severe patients compared to uncomplicated ones. linoleic acid oxylipins 231-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 27819189-0 2017 Inhibitory effects of curculigoside on human liver cytochrome P450 enzymes. curculigoside 22-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-66 27819189-2 2017 Curculigoside possesses numerous pharmacological activities, and however, little data available for the effects of curculigoside on the activity of human liver cytochrome P450 (CYP) enzymes. curculigoside 115-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-175 27819189-4 2017 This study investigates the inhibitory effects of curculigoside on the main human liver CYP isoforms. curculigoside 50-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 27819189-5 2017 In this study, the inhibitory effects of curculigoside on the eight human liver CYP isoforms 1A2, 2A6, 2E1, 2D6, 2C9, 2C19, 2C8, and 3A4 were investigated in human liver microsomes. curculigoside 41-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 27819189-7 2017 The results indicated that curculigoside could inhibit the activity of CYP1A2, CYP2C8, and CYP3A4, with IC50 values of 15.26, 11.93, and 9.47 muM, respectively, but that other CYP isoforms were not affected. curculigoside 27-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 27819189-11 2017 The in vitro studies of curculigoside with CYP isoforms suggest that curculigoside has the potential to cause pharmacokinetic drug interactions with other coadministered drugs metabolized by CYP1A2, CYP2C8, and CYP3A4. curculigoside 24-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 27819189-11 2017 The in vitro studies of curculigoside with CYP isoforms suggest that curculigoside has the potential to cause pharmacokinetic drug interactions with other coadministered drugs metabolized by CYP1A2, CYP2C8, and CYP3A4. curculigoside 69-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 28951593-1 2017 MicroRNAs (miRNAs) that regulate the cytochrome P-450 isoforms involved in acetaminophen (APAP) toxicity were examined in HepaRG cells treated with APAP (20 mM). Acetaminophen 75-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 28951593-1 2017 MicroRNAs (miRNAs) that regulate the cytochrome P-450 isoforms involved in acetaminophen (APAP) toxicity were examined in HepaRG cells treated with APAP (20 mM). Acetaminophen 90-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 28951593-1 2017 MicroRNAs (miRNAs) that regulate the cytochrome P-450 isoforms involved in acetaminophen (APAP) toxicity were examined in HepaRG cells treated with APAP (20 mM). Acetaminophen 148-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 28732807-7 2017 Moreover, the metabolism of alpha- and beta-asarone occurs mainly through cytochrome-P450 pathways. alpha- and beta-asarone 28-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 28370390-8 2017 Voriconazole-N-oxide inhibition of cytochrome P450 (CYP) isoforms 2C19 and 3A4 were assessed with the P450-Glo luminescence assay. Voriconazole N-Oxide 0-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 28370390-8 2017 Voriconazole-N-oxide inhibition of cytochrome P450 (CYP) isoforms 2C19 and 3A4 were assessed with the P450-Glo luminescence assay. Voriconazole N-Oxide 0-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 28370390-11 2017 Concentration giving 50% CYP inhibition of voriconazole N-oxide was 146 +- 23 mumol l-1 for CYP3A4, and 40.2 +- 4.2 mumol l-1 for CYP2C19. Voriconazole N-Oxide 43-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 28991531-0 2017 Identification of human cytochrome P450 and UGT enzymes involved in the metabolism of ferulic acid, a major bioactive component in traditional Chinese medicines. ferulic acid 86-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-47 28628909-0 2017 Nonalcoholic fatty liver disease impairs the cytochrome P-450-dependent metabolism of alpha-tocopherol (vitamin E). alpha-Tocopherol 86-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 28628909-0 2017 Nonalcoholic fatty liver disease impairs the cytochrome P-450-dependent metabolism of alpha-tocopherol (vitamin E). Vitamin E 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 28731962-1 2017 Cytochrome P450 oxidoreductase (POR) has played a potential role in the metabolism of drugs and steroids by supplying electrons to microsomal cytochrome P450 (CYP) enzymes. Steroids 96-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 28731962-1 2017 Cytochrome P450 oxidoreductase (POR) has played a potential role in the metabolism of drugs and steroids by supplying electrons to microsomal cytochrome P450 (CYP) enzymes. Steroids 96-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-162 28741929-1 2017 Cytochrome P450 CYP101A1 (P450cam) hydroxylates camphor by receiving two distinct electrons from its unique reductase, putidaredoxin (Pdx). Camphor 48-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28701464-1 2017 Recently, zebrafish and human cytochrome P450 (P450) 27C1 enzymes have been shown to be retinoid 3,4-desaturases. Retinoids 88-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-57 28495902-1 2017 A beta-blocker, metoprolol, is one of the in vivo probes for human cytochrome P450 (P450) 2D6. Metoprolol 16-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-93 27935260-7 2017 Studies using incubations with nicotinamide adenine dinucleotide phosphate-fortified pHLM with or without uridine 5"-diphosphoglucuronic acid and incubations with CYP-enzymes identified the main metabolic pathway of flubromazolam as hydroxylation on the alpha- and/or 4-position mediated by CYP3A4 and CYP3A5, with subsequent glucuronidation of the hydroxylated metabolites as well as of the parent drug. flubromazolam 216-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-166 29349890-2 2017 Potential atomoxetine (Strattera) and fluoxetine (Prozac) interactions via Cytochrome P450 (CYP450) pathways are examined and alternate therapies are recommended. Atomoxetine Hydrochloride 10-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 29349890-2 2017 Potential atomoxetine (Strattera) and fluoxetine (Prozac) interactions via Cytochrome P450 (CYP450) pathways are examined and alternate therapies are recommended. Atomoxetine Hydrochloride 10-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-98 29349890-2 2017 Potential atomoxetine (Strattera) and fluoxetine (Prozac) interactions via Cytochrome P450 (CYP450) pathways are examined and alternate therapies are recommended. Fluoxetine 38-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 29349890-2 2017 Potential atomoxetine (Strattera) and fluoxetine (Prozac) interactions via Cytochrome P450 (CYP450) pathways are examined and alternate therapies are recommended. Fluoxetine 38-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-98 29349890-2 2017 Potential atomoxetine (Strattera) and fluoxetine (Prozac) interactions via Cytochrome P450 (CYP450) pathways are examined and alternate therapies are recommended. Fluoxetine 50-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 29349890-2 2017 Potential atomoxetine (Strattera) and fluoxetine (Prozac) interactions via Cytochrome P450 (CYP450) pathways are examined and alternate therapies are recommended. Fluoxetine 50-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-98 27896399-0 2017 Cytochrome P450-mediated metabolism of triclosan attenuates its cytotoxicity in hepatic cells. Triclosan 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28726718-0 2017 Inhibitory Effects of Trapping Agents of Sulfur Drug Reactive Intermediates against Major Human Cytochrome P450 Isoforms. Sulfur 41-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-111 28726718-7 2017 To illustrate effects of CYP inhibition by trapping agents on reactive intermediate trapping, an example drug (ticlopidine) and trapping agent (NEM) were chosen for further studies. Ticlopidine 111-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 28726718-8 2017 For the same amount of ticlopidine (1 muM), increasing concentrations of the trapping agent NEM (0.007-40 mM) resulted in a bell-shaped response curve of NEM-trapped ticlopidine S-oxide (TSO-NEM), due to CYP inhibition by NEM. Ticlopidine S-oxide 166-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-207 28726718-8 2017 For the same amount of ticlopidine (1 muM), increasing concentrations of the trapping agent NEM (0.007-40 mM) resulted in a bell-shaped response curve of NEM-trapped ticlopidine S-oxide (TSO-NEM), due to CYP inhibition by NEM. tso-nem 187-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-207 28104494-5 2017 We review the synthesis and roles of especially PGRMC1- and cyP450-bound heme, the sources and transport of cholesterol, the involvement of PGRMC1 in cholesterol regulation, and the production of the first progestogen pregnenolone from cholesterol. Heme 73-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-66 28747995-1 2017 BACKGROUND: The objective of this study was to examine the inhibitory potential of darifenacin, fesoterodine, oxybutynin, propiverine, solifenacin, tolterodine and trospium chloride on the seven major human cytochrome P450 enzymes (CYP) by using a standardized and validated seven-in-one cytochrome P450 cocktail inhibition assay. trospium chloride 164-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-222 28747995-1 2017 BACKGROUND: The objective of this study was to examine the inhibitory potential of darifenacin, fesoterodine, oxybutynin, propiverine, solifenacin, tolterodine and trospium chloride on the seven major human cytochrome P450 enzymes (CYP) by using a standardized and validated seven-in-one cytochrome P450 cocktail inhibition assay. trospium chloride 164-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 288-303 28366651-0 2017 The impact of porous silicon nanoparticles on human cytochrome P450 metabolism in human liver microsomes in vitro. Silicon 21-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 28366651-2 2017 This study focuses on the hitherto unaddressed impact of porous silicon (PSi) nanoparticles on human cytochrome P450 (CYP) metabolism, which is the major detoxification route of most pharmaceuticals and other xenobiotics. Silicon 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 28366651-2 2017 This study focuses on the hitherto unaddressed impact of porous silicon (PSi) nanoparticles on human cytochrome P450 (CYP) metabolism, which is the major detoxification route of most pharmaceuticals and other xenobiotics. Silicon 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 27896399-8 2017 Consistent with the in vitro screening data, triclosan was extensively metabolized in HepG2 cells overexpressing CYP1A2, CYP2B6, CYP2C19, CYP2D6, and CYP2C18, and these cells were much more resistant to triclosan-induced cytotoxicity compared to vector cells, suggesting that CYP-mediated metabolism of triclosan attenuated its cytotoxicity. Triclosan 45-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 27896399-8 2017 Consistent with the in vitro screening data, triclosan was extensively metabolized in HepG2 cells overexpressing CYP1A2, CYP2B6, CYP2C19, CYP2D6, and CYP2C18, and these cells were much more resistant to triclosan-induced cytotoxicity compared to vector cells, suggesting that CYP-mediated metabolism of triclosan attenuated its cytotoxicity. Triclosan 203-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 27896399-8 2017 Consistent with the in vitro screening data, triclosan was extensively metabolized in HepG2 cells overexpressing CYP1A2, CYP2B6, CYP2C19, CYP2D6, and CYP2C18, and these cells were much more resistant to triclosan-induced cytotoxicity compared to vector cells, suggesting that CYP-mediated metabolism of triclosan attenuated its cytotoxicity. Triclosan 203-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 27896399-10 2017 Conjugation of triclosan, catalyzed by human glucuronosyltransferases (UGTs) and sulfotransferases (SULTs), also occurred in HepG2/CYP-overexpressing cells and primary human hepatocytes, with a greater extent of conjugation being associated with higher cell viability. Triclosan 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 27896399-12 2017 Among the 18 CYP-overexpressing cell lines, an inverse correlation was observed between cell viability and the level of triclosan in the culture medium. Triclosan 120-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 27896399-13 2017 In conclusion, human CYP isoforms that metabolize triclosan were identified, and the metabolism of triclosan by CYPs, UGTs, and SULTs decreased its cytotoxicity in hepatic cells. Triclosan 50-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 27896399-13 2017 In conclusion, human CYP isoforms that metabolize triclosan were identified, and the metabolism of triclosan by CYPs, UGTs, and SULTs decreased its cytotoxicity in hepatic cells. Triclosan 99-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 27896399-2 2017 The objectives of this study were to identify which cytochrome P450 (CYP) isoforms metabolize triclosan and to examine the effects of CYP-mediated metabolism on triclosan-induced cytotoxicity. Triclosan 94-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 27896399-2 2017 The objectives of this study were to identify which cytochrome P450 (CYP) isoforms metabolize triclosan and to examine the effects of CYP-mediated metabolism on triclosan-induced cytotoxicity. Triclosan 94-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 27896399-2 2017 The objectives of this study were to identify which cytochrome P450 (CYP) isoforms metabolize triclosan and to examine the effects of CYP-mediated metabolism on triclosan-induced cytotoxicity. Triclosan 161-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-137 27896399-4 2017 The extent of triclosan metabolism by each CYP was assessed by reversed-phase high-performance liquid chromatography with online radiochemical detection. Triclosan 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 28959657-0 2017 CYP polymorphisms and pathological conditions related to chronic exposure to organochlorine pesticides. Hydrocarbons, Chlorinated 77-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 28369648-3 2017 Cytochrome P450 enzymes are expressed at a relatively high level in astrocytes and may play a critical role in the biotransformation of endogenous or exogenous compounds, including chlorpyrifos, an organophosphate insecticide that affects the central nervous system. Chlorpyrifos 181-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28369648-3 2017 Cytochrome P450 enzymes are expressed at a relatively high level in astrocytes and may play a critical role in the biotransformation of endogenous or exogenous compounds, including chlorpyrifos, an organophosphate insecticide that affects the central nervous system. Organophosphates 198-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27412850-0 2017 In vitro metabolism of the anti-inflammatory clerodane diterpenoid polyandric acid A and its hydrolysis product by human liver microsomes and recombinant cytochrome P450 and UDP-glucuronosyltransferase enzymes. Diterpenes, Clerodane 45-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 27412850-0 2017 In vitro metabolism of the anti-inflammatory clerodane diterpenoid polyandric acid A and its hydrolysis product by human liver microsomes and recombinant cytochrome P450 and UDP-glucuronosyltransferase enzymes. Diterpenes 55-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 27412850-0 2017 In vitro metabolism of the anti-inflammatory clerodane diterpenoid polyandric acid A and its hydrolysis product by human liver microsomes and recombinant cytochrome P450 and UDP-glucuronosyltransferase enzymes. polyandric acid A 67-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. Diterpenes 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 344-359 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. Diterpenes 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 361-364 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. polyandric acid A 52-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 344-359 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. polyandric acid A 52-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 361-364 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. polyandric acid A 71-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 344-359 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. polyandric acid A 71-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 361-364 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. Alcohols 178-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 344-359 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. Alcohols 178-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 361-364 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. polyandric acid A 209-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 344-359 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. polyandric acid A 209-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 361-364 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. paah 228-232 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 344-359 27412850-2 2017 The metabolism of the anti-inflammatory diterpenoid polyandric acid A (PAA), a constituent of the Australian Aboriginal medicinal plant Dodonaea polyandra, and its de-esterified alcohol metabolite, hydrolysed polyandric acid A (PAAH) was studied in vitro using human liver microsomes (HLM) and recombinant UDP-glucuronosyltransferase (UGT) and cytochrome P450 (CYP) enzymes. paah 228-232 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 361-364 27412850-7 2017 Reaction phenotyping utilising selective enzyme inhibitors and recombinant human UGT and CYP enzymes revealed UGT2B7 and UGT1A1, and CYP2C9 and CYP3A4 as the major enzymes involved in the metabolism of PAAH. paah 202-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 28959657-1 2017 The association between genetic variations in the cytochrome P450 (CYP) family genes and pathological conditions related to long-term exposure to organochlorine compounds (OCs) deserves further elucidation. Hydrocarbons, Chlorinated 146-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 28959657-1 2017 The association between genetic variations in the cytochrome P450 (CYP) family genes and pathological conditions related to long-term exposure to organochlorine compounds (OCs) deserves further elucidation. Hydrocarbons, Chlorinated 146-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 28524079-8 2017 CYP450 activity was induced/inhibited in spheroids as expected, separate from any toxic response. spheroids 41-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 28300390-0 2017 Recruitment and Regulation of the Non-ribosomal Peptide Synthetase Modifying Cytochrome P450 Involved in Nikkomycin Biosynthesis. nikkomycin 105-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 28300390-2 2017 The cytochrome P450 (NikQ) hydroxylates the amino acid while it is appended via a phosphopantetheine linker to the non-ribosomal peptide synthetase (NRPS) NikP1. 4'-phosphopantetheine 82-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 28173639-0 2017 Bosentan and Rifampin Interactions Modulate Influx Transporter and Cytochrome P450 Expression and Activities in Primary Human Hepatocytes. Bosentan 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 28173639-0 2017 Bosentan and Rifampin Interactions Modulate Influx Transporter and Cytochrome P450 Expression and Activities in Primary Human Hepatocytes. Rifampin 13-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 28173639-3 2017 In this study, the effects of bosentan and rifampin on the expression and activities of organic anion-transporting peptide (OATP) and cytochrome P450 (CYP450) 2C9 and CYP3A4 were investigated in vitro. Bosentan 30-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-149 28173639-3 2017 In this study, the effects of bosentan and rifampin on the expression and activities of organic anion-transporting peptide (OATP) and cytochrome P450 (CYP450) 2C9 and CYP3A4 were investigated in vitro. Bosentan 30-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-157 28173639-3 2017 In this study, the effects of bosentan and rifampin on the expression and activities of organic anion-transporting peptide (OATP) and cytochrome P450 (CYP450) 2C9 and CYP3A4 were investigated in vitro. Rifampin 43-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-149 28173639-3 2017 In this study, the effects of bosentan and rifampin on the expression and activities of organic anion-transporting peptide (OATP) and cytochrome P450 (CYP450) 2C9 and CYP3A4 were investigated in vitro. Rifampin 43-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-157 28173639-7 2017 These results confirm rifampin- and bosentan-induced interactions between OATP transporters and CYP450. Rifampin 22-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-102 28173639-7 2017 These results confirm rifampin- and bosentan-induced interactions between OATP transporters and CYP450. Bosentan 36-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-102 28403941-14 2017 Since 5-HETE is a 5LOX product, 11-HETE is marker of lipid peroxidation, and vicinal diols are formed through soluble epoxide hydrolase (sEH) metabolism of CYP epoxygenated PUFAs, therefore, these results indicate that obesity is likely associated with altered metabolism with distinct oxygenating pathways. vicinal diols 77-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-159 28468305-2 2017 The inhibitory potentials of dimethyllirioresinol, epimagnolin A, eudesmin, fargesin, and magnolin on eight major human cytochrome P450 (CYP) enzyme activities in human liver microsomes were evaluated using liquid chromatography-tandem mass spectrometry to determine the inhibition mechanisms and inhibition potency. dimethyllirioresinol 29-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 28468305-2 2017 The inhibitory potentials of dimethyllirioresinol, epimagnolin A, eudesmin, fargesin, and magnolin on eight major human cytochrome P450 (CYP) enzyme activities in human liver microsomes were evaluated using liquid chromatography-tandem mass spectrometry to determine the inhibition mechanisms and inhibition potency. Epimagnolin A 51-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 28468305-2 2017 The inhibitory potentials of dimethyllirioresinol, epimagnolin A, eudesmin, fargesin, and magnolin on eight major human cytochrome P450 (CYP) enzyme activities in human liver microsomes were evaluated using liquid chromatography-tandem mass spectrometry to determine the inhibition mechanisms and inhibition potency. Epimagnolin A 51-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 28468305-2 2017 The inhibitory potentials of dimethyllirioresinol, epimagnolin A, eudesmin, fargesin, and magnolin on eight major human cytochrome P450 (CYP) enzyme activities in human liver microsomes were evaluated using liquid chromatography-tandem mass spectrometry to determine the inhibition mechanisms and inhibition potency. fargesin 76-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 28468305-0 2017 Inhibitory Effects of Dimethyllirioresinol, Epimagnolin A, Eudesmin, Fargesin, and Magnolin on Cytochrome P450 Enzyme Activities in Human Liver Microsomes. dimethyllirioresinol 22-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 28468305-0 2017 Inhibitory Effects of Dimethyllirioresinol, Epimagnolin A, Eudesmin, Fargesin, and Magnolin on Cytochrome P450 Enzyme Activities in Human Liver Microsomes. Epimagnolin A 44-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 28468305-0 2017 Inhibitory Effects of Dimethyllirioresinol, Epimagnolin A, Eudesmin, Fargesin, and Magnolin on Cytochrome P450 Enzyme Activities in Human Liver Microsomes. fargesin 69-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 28468305-0 2017 Inhibitory Effects of Dimethyllirioresinol, Epimagnolin A, Eudesmin, Fargesin, and Magnolin on Cytochrome P450 Enzyme Activities in Human Liver Microsomes. magnolin 83-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 28376352-2 2017 This study aims to identify the metabolic profile of five flavonoids (loureirin A, loureirin B, loureirin C, 7,4"-dihydroxyflavone and 5,7,4"-trihydroxyflavanone) from DB in HLMs as well as the CYP enzymes that are involved in the metabolism of them. Flavonoids 58-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-197 27226420-9 2017 Both cytochrome p450 (CYP) 3A4 and non-CYP mechanisms govern the clearance of panobinostat. Panobinostat 78-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-20 28301931-0 2017 Tunneling Effect That Changes the Reaction Pathway from Epoxidation to Hydroxylation in the Oxidation of Cyclohexene by a Compound I Model of Cytochrome P450. cyclohexene 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 27868228-9 2017 IMPLICATIONS FOR THERAPY: Amiodarone inhibits cytochrome P450 (CYP) isoforms 2C8 and 3A4 as well as P-glycoprotein (P-gp) for which taxanes are substrates. Amiodarone 26-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 27868228-9 2017 IMPLICATIONS FOR THERAPY: Amiodarone inhibits cytochrome P450 (CYP) isoforms 2C8 and 3A4 as well as P-glycoprotein (P-gp) for which taxanes are substrates. Amiodarone 26-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 27868228-9 2017 IMPLICATIONS FOR THERAPY: Amiodarone inhibits cytochrome P450 (CYP) isoforms 2C8 and 3A4 as well as P-glycoprotein (P-gp) for which taxanes are substrates. Taxoids 132-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 27868228-9 2017 IMPLICATIONS FOR THERAPY: Amiodarone inhibits cytochrome P450 (CYP) isoforms 2C8 and 3A4 as well as P-glycoprotein (P-gp) for which taxanes are substrates. Taxoids 132-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 28340451-3 2017 Drug-drug interactions (DDIs) may occur in prostate cancer patients due to inhibition by abiraterone of liver cytochrome P450 (CYP)-dependent enzymes CYP2C8 and 2D6, which are involved in the metabolism of approximately 25% of all drugs, and induction by enzalutamide of CYP3A4, 2C9 and 2C19, which metabolize up to 50% of medications. abiraterone 89-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 28340451-3 2017 Drug-drug interactions (DDIs) may occur in prostate cancer patients due to inhibition by abiraterone of liver cytochrome P450 (CYP)-dependent enzymes CYP2C8 and 2D6, which are involved in the metabolism of approximately 25% of all drugs, and induction by enzalutamide of CYP3A4, 2C9 and 2C19, which metabolize up to 50% of medications. abiraterone 89-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 28340451-3 2017 Drug-drug interactions (DDIs) may occur in prostate cancer patients due to inhibition by abiraterone of liver cytochrome P450 (CYP)-dependent enzymes CYP2C8 and 2D6, which are involved in the metabolism of approximately 25% of all drugs, and induction by enzalutamide of CYP3A4, 2C9 and 2C19, which metabolize up to 50% of medications. enzalutamide 255-267 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 28340451-3 2017 Drug-drug interactions (DDIs) may occur in prostate cancer patients due to inhibition by abiraterone of liver cytochrome P450 (CYP)-dependent enzymes CYP2C8 and 2D6, which are involved in the metabolism of approximately 25% of all drugs, and induction by enzalutamide of CYP3A4, 2C9 and 2C19, which metabolize up to 50% of medications. enzalutamide 255-267 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 28274629-0 2017 Effects of 6-paradol, an unsaturated ketone from gingers, on cytochrome P450-mediated drug metabolism. 6-paradol 11-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 28274629-0 2017 Effects of 6-paradol, an unsaturated ketone from gingers, on cytochrome P450-mediated drug metabolism. unsaturated ketone 25-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 28274629-3 2017 In this study, the inhibitory effects of 6-paradol on the activities of cytochrome P450 (CYP) enzymes were investigated with human liver microsomes and recombinant CYP isozymes. 6-paradol 41-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 28274629-3 2017 In this study, the inhibitory effects of 6-paradol on the activities of cytochrome P450 (CYP) enzymes were investigated with human liver microsomes and recombinant CYP isozymes. 6-paradol 41-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 28274629-3 2017 In this study, the inhibitory effects of 6-paradol on the activities of cytochrome P450 (CYP) enzymes were investigated with human liver microsomes and recombinant CYP isozymes. 6-paradol 41-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-167 28274629-4 2017 6-Paradol showed concentration-dependent inhibitory effects on CYP1A2, CYP2B6, CYP2C8, CYP2C9, and CYP2C19 isozymes, with IC50 values ranging from 3.8 to 21.4microM in recombinant CYP isozymes. 6-paradol 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 27449533-2 2017 CyP4F22, a protein of the cytochrome-P450 family 4, encodes an epidermal omega-hydroxylase decisive in the formation of acylceramides, which is hypothesized to be crucial for skin-barrier function. acylceramides 120-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-50 27347703-11 2017 These results demonstrate that luseogliflozin is metabolized through multiple pathways, including CYP-mediated oxidation and glucuronidation, in human. 1,5-anhydro-1-(5-(4-ethoxybenzyl)-2-methoxy-4-methylphenyl)-1-thioglucitol 31-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 27226420-9 2017 Both cytochrome p450 (CYP) 3A4 and non-CYP mechanisms govern the clearance of panobinostat. Panobinostat 78-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 27226420-9 2017 Both cytochrome p450 (CYP) 3A4 and non-CYP mechanisms govern the clearance of panobinostat. Panobinostat 78-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 28377720-1 2017 Cytochrome P450 enzymes are often responsible for the toxic and carcinogenic effects of toxicants, such as aflatoxin B1 (AFB1). Aflatoxin B1 107-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28215634-8 2017 We have confirmed that the hepatocyte-like cells prepared by our methods were able to increase gene expression of cytochrome P450 enzymes upon encountering rifampicin, phenobarbital, or omeprazole. Rifampin 156-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 28215634-8 2017 We have confirmed that the hepatocyte-like cells prepared by our methods were able to increase gene expression of cytochrome P450 enzymes upon encountering rifampicin, phenobarbital, or omeprazole. Phenobarbital 168-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 28215634-8 2017 We have confirmed that the hepatocyte-like cells prepared by our methods were able to increase gene expression of cytochrome P450 enzymes upon encountering rifampicin, phenobarbital, or omeprazole. Omeprazole 186-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 28377720-1 2017 Cytochrome P450 enzymes are often responsible for the toxic and carcinogenic effects of toxicants, such as aflatoxin B1 (AFB1). Aflatoxin B1 121-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28331291-5 2017 The effect of genetic polymorphisms of cytochrome-P450 (CYP) enzymes on the pharmacokinetics of DA-8031 was evaluated. DA 8031 96-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 28331291-5 2017 The effect of genetic polymorphisms of cytochrome-P450 (CYP) enzymes on the pharmacokinetics of DA-8031 was evaluated. DA 8031 96-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 28287454-0 2017 AM-2201 Inhibits Multiple Cytochrome P450 and Uridine 5"-Diphospho-Glucuronosyltransferase Enzyme Activities in Human Liver Microsomes. 1-(5-fluoropentyl)-3-(1-naphthoyl)indole 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 27718269-0 2017 Characterization of human cytochrome P450 mediated bioactivation of amodiaquine and its major metabolite N-desethylamodiaquine. Amodiaquine 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 28287454-2 2017 However, there are no reports of the inhibitory effect of AM-2201 on human cytochrome P450 (CYP) or uridine 5"-diphospho-glucuronosyltransferase (UGT) enzymes. 1-(5-fluoropentyl)-3-(1-naphthoyl)indole 58-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 27718269-0 2017 Characterization of human cytochrome P450 mediated bioactivation of amodiaquine and its major metabolite N-desethylamodiaquine. desethylamodiaquine 105-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 27718269-1 2017 AIMS: Oxidative bioactivation of amodiaquine (AQ) by cytochrome P450s to a reactive quinoneimine is considered as an important mechanism underlying its idiosyncratic hepatotoxicity. Amodiaquine 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 27718269-1 2017 AIMS: Oxidative bioactivation of amodiaquine (AQ) by cytochrome P450s to a reactive quinoneimine is considered as an important mechanism underlying its idiosyncratic hepatotoxicity. Amodiaquine 46-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 27718269-1 2017 AIMS: Oxidative bioactivation of amodiaquine (AQ) by cytochrome P450s to a reactive quinoneimine is considered as an important mechanism underlying its idiosyncratic hepatotoxicity. quinoneimine 84-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 27924364-0 2017 Metabolic characterization of (1-(5-fluoropentyl)-1H-indol-3-yl)(4-methyl-1-naphthalenyl)-methanone (MAM-2201) using human liver microsomes and cDNA-overexpressed cytochrome P450 enzymes. 1-(5-fluoropentyl)-1h-indol-3-yl) 31-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-178 28253085-3 2017 To the best of our knowledge, no study to date has evaluated the effects of water pipe smoking on cytochrome P450 (CYP450) activities and drug interaction potential in humans, whereas only limited information is available on the impact of secondhand smoke on drug metabolism. Water 76-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 28253085-11 2017 Further studies in larger prospective samples are recommended to evaluate water pipe and secondhand tobacco smoke effects on CYP450 function, particularly at higher smoke exposure conditions. Water 74-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-131 28140586-3 2017 These also are often found near cytochrome P450 (CYP) mono-oxygenases that presumably further decorate the ensuing diterpene, suggesting that these fungi might produce more elaborate diterpenoids. Diterpenes 115-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 28140586-3 2017 These also are often found near cytochrome P450 (CYP) mono-oxygenases that presumably further decorate the ensuing diterpene, suggesting that these fungi might produce more elaborate diterpenoids. Diterpenes 115-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 28140586-3 2017 These also are often found near cytochrome P450 (CYP) mono-oxygenases that presumably further decorate the ensuing diterpene, suggesting that these fungi might produce more elaborate diterpenoids. Diterpenes 183-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 28140586-3 2017 These also are often found near cytochrome P450 (CYP) mono-oxygenases that presumably further decorate the ensuing diterpene, suggesting that these fungi might produce more elaborate diterpenoids. Diterpenes 183-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 27966929-2 2017 Experimental studies indicate that cytochrome P450 (CYP) enzymes catalyze the bioactivation of estrogens to catechols, which can exert their harmful effects via various routes. Catechols 108-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 27966929-2 2017 Experimental studies indicate that cytochrome P450 (CYP) enzymes catalyze the bioactivation of estrogens to catechols, which can exert their harmful effects via various routes. Catechols 108-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 27966929-6 2017 We studied the structure of human estrogen metabolizing enzymes (CYP1A1, CYP1A2, CYP1B1, and CYP3A4) in complex with estrone using docking and investigated the susceptibility of estrone, equilin, and equilenin (which only differ in the unsaturation of ring B) to undergo 2- and 4-hydroxylation using several models of CYP enzymes (Compound I, methoxy, and phenoxy radical). Estrone 117-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 27966929-6 2017 We studied the structure of human estrogen metabolizing enzymes (CYP1A1, CYP1A2, CYP1B1, and CYP3A4) in complex with estrone using docking and investigated the susceptibility of estrone, equilin, and equilenin (which only differ in the unsaturation of ring B) to undergo 2- and 4-hydroxylation using several models of CYP enzymes (Compound I, methoxy, and phenoxy radical). Estrone 178-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 27966929-6 2017 We studied the structure of human estrogen metabolizing enzymes (CYP1A1, CYP1A2, CYP1B1, and CYP3A4) in complex with estrone using docking and investigated the susceptibility of estrone, equilin, and equilenin (which only differ in the unsaturation of ring B) to undergo 2- and 4-hydroxylation using several models of CYP enzymes (Compound I, methoxy, and phenoxy radical). Equilin 187-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 27966929-6 2017 We studied the structure of human estrogen metabolizing enzymes (CYP1A1, CYP1A2, CYP1B1, and CYP3A4) in complex with estrone using docking and investigated the susceptibility of estrone, equilin, and equilenin (which only differ in the unsaturation of ring B) to undergo 2- and 4-hydroxylation using several models of CYP enzymes (Compound I, methoxy, and phenoxy radical). Equilenin 200-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 27966929-6 2017 We studied the structure of human estrogen metabolizing enzymes (CYP1A1, CYP1A2, CYP1B1, and CYP3A4) in complex with estrone using docking and investigated the susceptibility of estrone, equilin, and equilenin (which only differ in the unsaturation of ring B) to undergo 2- and 4-hydroxylation using several models of CYP enzymes (Compound I, methoxy, and phenoxy radical). 7-methoxy-6-(2'-methoxy-3'-hydroxy-3'-methyl butyl) 343-350 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 27966929-6 2017 We studied the structure of human estrogen metabolizing enzymes (CYP1A1, CYP1A2, CYP1B1, and CYP3A4) in complex with estrone using docking and investigated the susceptibility of estrone, equilin, and equilenin (which only differ in the unsaturation of ring B) to undergo 2- and 4-hydroxylation using several models of CYP enzymes (Compound I, methoxy, and phenoxy radical). phenoxy radical 356-371 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 28080030-1 2017 Cytochrome P450 enzymes have been engineered to catalyze abiological C-H bond amination reactions, but the yields of these reactions have been limited by low chemoselectivity for the amination of C-H bonds over competing reduction of the azide substrate to a sulfonamide. Azides 238-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28080030-1 2017 Cytochrome P450 enzymes have been engineered to catalyze abiological C-H bond amination reactions, but the yields of these reactions have been limited by low chemoselectivity for the amination of C-H bonds over competing reduction of the azide substrate to a sulfonamide. Sulfonamides 259-270 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27457692-5 2017 AIM OF THE STUDY: Aim of the Study was to investigate herb-drug interaction (HDI) of Ridayarishta formulation through human hepatic cytochrome P450 (CYP450) enzyme inhibition assay. ridayarishta 85-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-147 27457692-5 2017 AIM OF THE STUDY: Aim of the Study was to investigate herb-drug interaction (HDI) of Ridayarishta formulation through human hepatic cytochrome P450 (CYP450) enzyme inhibition assay. ridayarishta 85-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-155 27457692-14 2017 It may be concluded that consumption of Ridayarishta along with selective cardio protective, antihypertensive and anti-diabetic conventional medicine is safe with negligible or without any significant CYP450 (CYP1A2, 2C19, 2D6 and 3A4) inhibition mediated HDI. ridayarishta 40-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-207 28344492-3 2017 Cytochrome P450 (CYP) enzymes, an essential source of variability in drug-response, play role in not only phase I-dependent metabolism of xenobiotics but also metabolism of endogenous compounds such as steroids, vitamins and fatty acids. Steroids 202-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28344492-3 2017 Cytochrome P450 (CYP) enzymes, an essential source of variability in drug-response, play role in not only phase I-dependent metabolism of xenobiotics but also metabolism of endogenous compounds such as steroids, vitamins and fatty acids. Steroids 202-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 28344492-3 2017 Cytochrome P450 (CYP) enzymes, an essential source of variability in drug-response, play role in not only phase I-dependent metabolism of xenobiotics but also metabolism of endogenous compounds such as steroids, vitamins and fatty acids. Fatty Acids 225-236 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28344492-3 2017 Cytochrome P450 (CYP) enzymes, an essential source of variability in drug-response, play role in not only phase I-dependent metabolism of xenobiotics but also metabolism of endogenous compounds such as steroids, vitamins and fatty acids. Fatty Acids 225-236 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 27924364-0 2017 Metabolic characterization of (1-(5-fluoropentyl)-1H-indol-3-yl)(4-methyl-1-naphthalenyl)-methanone (MAM-2201) using human liver microsomes and cDNA-overexpressed cytochrome P450 enzymes. 4-methyl-1-naphthalenyl)-methanone 65-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-178 26874845-0 2017 Inhibitory Effects of Triptolide on Human Liver Cytochrome P450 Enzymes and P-Glycoprotein. triptolide 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 26874845-2 2017 However, it remains unclear how triptolide influences the activity of human liver cytochrome P450 (CYP) enzymes and P-glycoprotein (P-gp). triptolide 32-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 26874845-2 2017 However, it remains unclear how triptolide influences the activity of human liver cytochrome P450 (CYP) enzymes and P-glycoprotein (P-gp). triptolide 32-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-102 26874845-3 2017 METHODS: In this study, the inhibitory effects of triptolide on the eight human liver CYP isoforms (i.e., 1A2, 3A4, 2A6, 2E1, 2D6, 2C9, 2C19, and 2C8) were investigated in vitro using human liver microsomes (HLMs), and the effects of triptolide on the activity of P-gp were investigated using a rhodamine-123 uptake assay. triptolide 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 26874845-4 2017 RESULTS: The results showed that triptolide inhibited the activity of CYP1A2 and CYP3A4, with 50 % inhibitory concentration (IC50) values of 14.18 and 8.36 muM, respectively, but that other CYP isoforms were not affected. triptolide 33-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 26874845-8 2017 CONCLUSIONS: The in vitro studies of triptolide with CYP isoforms and P-gp indicate that triptolide has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP1A2 and CYP3A4. triptolide 37-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 26874845-8 2017 CONCLUSIONS: The in vitro studies of triptolide with CYP isoforms and P-gp indicate that triptolide has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP1A2 and CYP3A4. triptolide 89-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 28007573-1 2017 OBJECTIVE: Prominent among the endothelium-derived hyperpolarizing factors (EDHFs) are the Cytochrome P450 (CYP) epoxygenase-derived arachidonic acid metabolites-the epoxyeicosatrienoic acids (EETs), that are known as vasodilators in the microcirculation. edhfs 76-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 28007573-1 2017 OBJECTIVE: Prominent among the endothelium-derived hyperpolarizing factors (EDHFs) are the Cytochrome P450 (CYP) epoxygenase-derived arachidonic acid metabolites-the epoxyeicosatrienoic acids (EETs), that are known as vasodilators in the microcirculation. edhfs 76-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 28007573-1 2017 OBJECTIVE: Prominent among the endothelium-derived hyperpolarizing factors (EDHFs) are the Cytochrome P450 (CYP) epoxygenase-derived arachidonic acid metabolites-the epoxyeicosatrienoic acids (EETs), that are known as vasodilators in the microcirculation. Arachidonic Acid 133-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 28007573-1 2017 OBJECTIVE: Prominent among the endothelium-derived hyperpolarizing factors (EDHFs) are the Cytochrome P450 (CYP) epoxygenase-derived arachidonic acid metabolites-the epoxyeicosatrienoic acids (EETs), that are known as vasodilators in the microcirculation. Arachidonic Acid 133-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 28007573-1 2017 OBJECTIVE: Prominent among the endothelium-derived hyperpolarizing factors (EDHFs) are the Cytochrome P450 (CYP) epoxygenase-derived arachidonic acid metabolites-the epoxyeicosatrienoic acids (EETs), that are known as vasodilators in the microcirculation. epoxyeicosatrienoic acids 166-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 28007573-1 2017 OBJECTIVE: Prominent among the endothelium-derived hyperpolarizing factors (EDHFs) are the Cytochrome P450 (CYP) epoxygenase-derived arachidonic acid metabolites-the epoxyeicosatrienoic acids (EETs), that are known as vasodilators in the microcirculation. epoxyeicosatrienoic acids 166-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 28007573-1 2017 OBJECTIVE: Prominent among the endothelium-derived hyperpolarizing factors (EDHFs) are the Cytochrome P450 (CYP) epoxygenase-derived arachidonic acid metabolites-the epoxyeicosatrienoic acids (EETs), that are known as vasodilators in the microcirculation. eets 193-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 28007573-1 2017 OBJECTIVE: Prominent among the endothelium-derived hyperpolarizing factors (EDHFs) are the Cytochrome P450 (CYP) epoxygenase-derived arachidonic acid metabolites-the epoxyeicosatrienoic acids (EETs), that are known as vasodilators in the microcirculation. eets 193-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 27861439-3 2017 The opioid analgesic potency of a given dose of tramadol is influenced by an individual"s CYP genetics, with poor metabolizers experiencing little conversion to the active M1 opioid metabolite and individuals with a high metabolic profile, or ultra-metabolizers, experiencing the greatest opioid analgesic effects. Tramadol 48-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-93 27712037-4 2017 The pharmacokinetics of CYP selective substrates: caffeine, losartan, and omeprazole changed significantly in a diabetic NASH mouse model, indicating attenuation of the activity of Cyp1a2 and Cyp2c29, respectively. Caffeine 50-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 27712037-4 2017 The pharmacokinetics of CYP selective substrates: caffeine, losartan, and omeprazole changed significantly in a diabetic NASH mouse model, indicating attenuation of the activity of Cyp1a2 and Cyp2c29, respectively. Losartan 60-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 27712037-4 2017 The pharmacokinetics of CYP selective substrates: caffeine, losartan, and omeprazole changed significantly in a diabetic NASH mouse model, indicating attenuation of the activity of Cyp1a2 and Cyp2c29, respectively. Omeprazole 74-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 28197402-0 2017 Suicide Inhibition of Cytochrome P450 Enzymes by Cyclopropylamines via a Ring-Opening Mechanism: Proton-Coupled Electron Transfer Makes a Difference. cyclopropylamine 49-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 28197402-1 2017 N-benzyl-N-cyclopropylamine (BCA) has been attracting great interests for decades for its partial suicide inactivation role to cytochrome P450 (P450) via a ring-opening mechanism besides acting as a role of normal substrates. N-Benzylcyclopropylamine 0-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-142 28197402-1 2017 N-benzyl-N-cyclopropylamine (BCA) has been attracting great interests for decades for its partial suicide inactivation role to cytochrome P450 (P450) via a ring-opening mechanism besides acting as a role of normal substrates. alpha-bromocinnamaldehyde 29-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-142 27861439-4 2017 The importance of the CYP metabolism has led to the adoption of computer clinical decision support with pharmacogenomics tools guiding tramadol treatment in major medical centers. Tramadol 135-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 27861439-7 2017 This review provides updated important information on the pharmacology, pharmacokinetics, CYP genetic polymorphisms, drug interactions, toxicity, withdrawal, and illicit use of tramadol. Tramadol 177-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-93 27273149-0 2017 Pharmacokinetic Effects of Isavuconazole Coadministration With the Cytochrome P450 Enzyme Substrates Bupropion, Repaglinide, Caffeine, Dextromethorphan, and Methadone in Healthy Subjects. isavuconazole 27-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 27482943-0 2017 The Pharmacogenomic Association of Fcgamma Receptors and Cytochrome P450 Enzymes With Response to Rituximab or Cyclophosphamide Treatment in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Cyclophosphamide 111-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 27273149-0 2017 Pharmacokinetic Effects of Isavuconazole Coadministration With the Cytochrome P450 Enzyme Substrates Bupropion, Repaglinide, Caffeine, Dextromethorphan, and Methadone in Healthy Subjects. Bupropion 101-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 27273149-0 2017 Pharmacokinetic Effects of Isavuconazole Coadministration With the Cytochrome P450 Enzyme Substrates Bupropion, Repaglinide, Caffeine, Dextromethorphan, and Methadone in Healthy Subjects. repaglinide 112-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 27273149-0 2017 Pharmacokinetic Effects of Isavuconazole Coadministration With the Cytochrome P450 Enzyme Substrates Bupropion, Repaglinide, Caffeine, Dextromethorphan, and Methadone in Healthy Subjects. Caffeine 125-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 27273149-0 2017 Pharmacokinetic Effects of Isavuconazole Coadministration With the Cytochrome P450 Enzyme Substrates Bupropion, Repaglinide, Caffeine, Dextromethorphan, and Methadone in Healthy Subjects. Dextromethorphan 135-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 27273149-0 2017 Pharmacokinetic Effects of Isavuconazole Coadministration With the Cytochrome P450 Enzyme Substrates Bupropion, Repaglinide, Caffeine, Dextromethorphan, and Methadone in Healthy Subjects. Methadone 157-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 27863990-0 2017 Cytochrome P450 enzymes but not NADPH oxidases are the source of the NADPH-dependent lucigenin chemiluminescence in membrane assays. 10,10'-dimethyl-9,9'-biacridinium 85-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27816611-3 2017 In the same years the cytochrome P-450 dependent metabolism of vitamin E was characterized and a first series of studies on short-chain carboxyethyl metabolites in the 1990s paved the way to the hypothesis of a biological role for this metabolism alternative to vitamin E catabolism. Vitamin E 63-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 27816611-4 2017 In the last decade other physiological metabolites of vitamin E have been identified, such as alpha-tocopheryl phosphate and the long-chain metabolites formed by the omega-hydroxylase activity of cytochrome P-450. Vitamin E 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 196-212 27863990-7 2017 Using a combination of native protein electrophoresis, NADPH-stimulated assays and mass spectrometry, mitochondrial proteins and cytochrome P450 were identified as possible source of the assay signal. NADP 55-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-144 29104319-1 2017 Abstract: Ellipticine is an anticancer agent that forms covalent DNA adducts after enzymatic activation by cytochrome P450 (CYP) enzymes, mainly by CYP3A4. ellipticine 10-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 29104319-1 2017 Abstract: Ellipticine is an anticancer agent that forms covalent DNA adducts after enzymatic activation by cytochrome P450 (CYP) enzymes, mainly by CYP3A4. ellipticine 10-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 29104318-7 2017 We found that in contrast to AAI, AAII is oxidized only by several CYP enzymatic systems and their efficiency is much lower for oxidation of AAII than AAI. aaii 34-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 28102631-0 2017 Cytochrome P450 isoform genotyping in poor propranolol responders for hemangioma treatment. Propranolol 43-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 29104318-7 2017 We found that in contrast to AAI, AAII is oxidized only by several CYP enzymatic systems and their efficiency is much lower for oxidation of AAII than AAI. aai 34-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 28132967-9 2017 Blood pressure control is mediated at least in part by cytochrome P450 (CYP) enzymes such as CYP4A11 and CYP4F2 that produce 20-hydroxyeicosatetraenoic acid (20-HETE), involved in salt balance in the kidney, and all are known to be altered during Cd exposure. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 125-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 28132967-9 2017 Blood pressure control is mediated at least in part by cytochrome P450 (CYP) enzymes such as CYP4A11 and CYP4F2 that produce 20-hydroxyeicosatetraenoic acid (20-HETE), involved in salt balance in the kidney, and all are known to be altered during Cd exposure. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 125-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 28132967-9 2017 Blood pressure control is mediated at least in part by cytochrome P450 (CYP) enzymes such as CYP4A11 and CYP4F2 that produce 20-hydroxyeicosatetraenoic acid (20-HETE), involved in salt balance in the kidney, and all are known to be altered during Cd exposure. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 158-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 28132967-9 2017 Blood pressure control is mediated at least in part by cytochrome P450 (CYP) enzymes such as CYP4A11 and CYP4F2 that produce 20-hydroxyeicosatetraenoic acid (20-HETE), involved in salt balance in the kidney, and all are known to be altered during Cd exposure. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 158-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 28132967-9 2017 Blood pressure control is mediated at least in part by cytochrome P450 (CYP) enzymes such as CYP4A11 and CYP4F2 that produce 20-hydroxyeicosatetraenoic acid (20-HETE), involved in salt balance in the kidney, and all are known to be altered during Cd exposure. Cadmium 247-249 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 28132967-9 2017 Blood pressure control is mediated at least in part by cytochrome P450 (CYP) enzymes such as CYP4A11 and CYP4F2 that produce 20-hydroxyeicosatetraenoic acid (20-HETE), involved in salt balance in the kidney, and all are known to be altered during Cd exposure. Cadmium 247-249 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 27452206-3 2016 Dimerization is required for association with cytochrome P450 (cyP450) enzymes, which mediates chemoresistance to doxorubicin and may be responsible for PGRMC1"s anti-apoptotic activity. Doxorubicin 114-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 27727524-1 2016 Cytochrome P450 enzymes are heme-containing mono-oxygenases that mainly react through oxygen-atom transfer. Oxygen 49-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 28006925-1 2016 This review summarizes the importance of bile acids (BA) as important regulators of various homeostatic mechanisms with detailed focus on cytochrome P450 (CYP) enzymes. Bile Acids and Salts 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-158 28006925-1 2016 This review summarizes the importance of bile acids (BA) as important regulators of various homeostatic mechanisms with detailed focus on cytochrome P450 (CYP) enzymes. Bile Acids and Salts 53-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-158 27452206-3 2016 Dimerization is required for association with cytochrome P450 (cyP450) enzymes, which mediates chemoresistance to doxorubicin and may be responsible for PGRMC1"s anti-apoptotic activity. Doxorubicin 114-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-69 27836712-0 2016 Application of substrate depletion assay to evaluation of CYP isoforms responsible for stereoselective metabolism of carvedilol. Carvedilol 117-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 27317413-2 2016 The majority of DAAs are extensively metabolized by liver enzymes and have the ability to influence cytochrome P450 (CYP) enzymes. daas 16-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 27317413-2 2016 The majority of DAAs are extensively metabolized by liver enzymes and have the ability to influence cytochrome P450 (CYP) enzymes. daas 16-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-120 27718490-3 2016 METHODS: To overcome this drawback, the cross-reactivities exhibited by six chemical inhibitors (furafylline, montelukast, sulfaphenazole, ticlopidine, quinidine and ketoconazole) were quantified using specific CYP enzyme marker reactions. Ketoconazole 166-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-214 27836712-1 2016 To evaluate the relative contribution of cytochrome P450 (CYP) isoforms responsible for carvedilol (CAR) oxidation, enantioselective metabolism of CAR was investigated in human liver microsomes (HLMs) and recombinant human CYPs by using the substrate depletion assay. Carvedilol 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 27836712-1 2016 To evaluate the relative contribution of cytochrome P450 (CYP) isoforms responsible for carvedilol (CAR) oxidation, enantioselective metabolism of CAR was investigated in human liver microsomes (HLMs) and recombinant human CYPs by using the substrate depletion assay. Carvedilol 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 27507240-9 2016 Pre-treatment with the cytochrome-P450 inhibitor malathion supports a non-target site metabolism-based mechanism of herbicide resistance. Malathion 49-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 27198207-1 2016 Tamoxifen is metabolically activated to 4-hydroxytamoxifen and endoxifen by cytochrome P450 (CYP). Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 27198207-1 2016 Tamoxifen is metabolically activated to 4-hydroxytamoxifen and endoxifen by cytochrome P450 (CYP). Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 27198207-1 2016 Tamoxifen is metabolically activated to 4-hydroxytamoxifen and endoxifen by cytochrome P450 (CYP). hydroxytamoxifen 40-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 27198207-1 2016 Tamoxifen is metabolically activated to 4-hydroxytamoxifen and endoxifen by cytochrome P450 (CYP). 4-hydroxy-N-desmethyltamoxifen 63-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 27198207-1 2016 Tamoxifen is metabolically activated to 4-hydroxytamoxifen and endoxifen by cytochrome P450 (CYP). 4-hydroxy-N-desmethyltamoxifen 63-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 27198207-2 2016 CYP phenotypes have been correlated to tamoxifen outcomes, but few have considered drug interactions or combinations of genes. Tamoxifen 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 27198207-11 2016 With 3 impaired CYP phenotypes, endoxifen concentrations were lower than if only CYP2D6 was impaired (P = .05). 4-hydroxy-N-desmethyltamoxifen 32-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 27779894-0 2016 Influence of Sulforaphane Metabolites on Activities of Human Drug-Metabolizing Cytochrome P450 and Determination of Sulforaphane in Human Liver Cells. sulforaphane 13-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 27289043-1 2016 The importance of Cytochrome P450-catalyzed modifications of natural products produced by non-ribosomal peptide synthetase machineries is most apparent during glycopeptide antibiotic biosynthesis: specifically, the formation of essential amino acid side chains crosslinks in the peptide backbone of these clinically relevant antibiotics. Amino Acids, Essential 228-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 27805325-0 2016 Corrigendum: Mutating a Highly Conserved Residue in Diverse Cytochrome P450s Facilitates Diastereoselective Olefin Cyclopropanation. Alkenes 108-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 29908557-0 2016 [The in vitro inhibition and induction of cytochrome P450 activities by bentysrepinine: a novel candidate of anti-hepatitis B virus drug]. N-(N-benzoyl-O-(2-dimethylaminoethyl)tyrosyl)phenylalaninol 72-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 26975734-1 2016 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to generate epoxyeicosatrienoic acids (EETs). Arachidonic Acid 46-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27591243-1 2016 Epoxyeicosatrienoic acids (EETs) are metabolic products of free arachidonic acid, which are produced through cytochrome P-450 (CYP) epoxygenases. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 27591243-1 2016 Epoxyeicosatrienoic acids (EETs) are metabolic products of free arachidonic acid, which are produced through cytochrome P-450 (CYP) epoxygenases. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 27591243-1 2016 Epoxyeicosatrienoic acids (EETs) are metabolic products of free arachidonic acid, which are produced through cytochrome P-450 (CYP) epoxygenases. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 27591243-1 2016 Epoxyeicosatrienoic acids (EETs) are metabolic products of free arachidonic acid, which are produced through cytochrome P-450 (CYP) epoxygenases. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 27591243-1 2016 Epoxyeicosatrienoic acids (EETs) are metabolic products of free arachidonic acid, which are produced through cytochrome P-450 (CYP) epoxygenases. Arachidonic Acid 64-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 27591243-1 2016 Epoxyeicosatrienoic acids (EETs) are metabolic products of free arachidonic acid, which are produced through cytochrome P-450 (CYP) epoxygenases. Arachidonic Acid 64-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 27087514-1 2016 AIM: To better understand the relationship between the interactions among rs17110453, rs751141, and rs9333025 variants and plasma levels of cytochrome P450 (CYP) metabolites, i.e.,20-hydroxyeicosatetraenoic acid (20-HETE), epoxyeicosatrienoic acids (EETs), and dihydroxyeicosatrienoic acids (DiHETEs) in ischemia stroke (IS). dihydroxyeicosatrienoic acids 261-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 27087514-1 2016 AIM: To better understand the relationship between the interactions among rs17110453, rs751141, and rs9333025 variants and plasma levels of cytochrome P450 (CYP) metabolites, i.e.,20-hydroxyeicosatetraenoic acid (20-HETE), epoxyeicosatrienoic acids (EETs), and dihydroxyeicosatrienoic acids (DiHETEs) in ischemia stroke (IS). dihetes 292-299 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 27087514-1 2016 AIM: To better understand the relationship between the interactions among rs17110453, rs751141, and rs9333025 variants and plasma levels of cytochrome P450 (CYP) metabolites, i.e.,20-hydroxyeicosatetraenoic acid (20-HETE), epoxyeicosatrienoic acids (EETs), and dihydroxyeicosatrienoic acids (DiHETEs) in ischemia stroke (IS). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 180-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-155 27087514-1 2016 AIM: To better understand the relationship between the interactions among rs17110453, rs751141, and rs9333025 variants and plasma levels of cytochrome P450 (CYP) metabolites, i.e.,20-hydroxyeicosatetraenoic acid (20-HETE), epoxyeicosatrienoic acids (EETs), and dihydroxyeicosatrienoic acids (DiHETEs) in ischemia stroke (IS). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 180-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 27087514-1 2016 AIM: To better understand the relationship between the interactions among rs17110453, rs751141, and rs9333025 variants and plasma levels of cytochrome P450 (CYP) metabolites, i.e.,20-hydroxyeicosatetraenoic acid (20-HETE), epoxyeicosatrienoic acids (EETs), and dihydroxyeicosatrienoic acids (DiHETEs) in ischemia stroke (IS). 20-Hete 213-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 27087514-1 2016 AIM: To better understand the relationship between the interactions among rs17110453, rs751141, and rs9333025 variants and plasma levels of cytochrome P450 (CYP) metabolites, i.e.,20-hydroxyeicosatetraenoic acid (20-HETE), epoxyeicosatrienoic acids (EETs), and dihydroxyeicosatrienoic acids (DiHETEs) in ischemia stroke (IS). epoxyeicosatrienoic acids 223-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 27087514-1 2016 AIM: To better understand the relationship between the interactions among rs17110453, rs751141, and rs9333025 variants and plasma levels of cytochrome P450 (CYP) metabolites, i.e.,20-hydroxyeicosatetraenoic acid (20-HETE), epoxyeicosatrienoic acids (EETs), and dihydroxyeicosatrienoic acids (DiHETEs) in ischemia stroke (IS). eets 250-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 26975734-0 2016 The factor in EDHF: Cytochrome P450 derived lipid mediators and vascular signaling. edhf 14-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 26975734-1 2016 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to generate epoxyeicosatrienoic acids (EETs). Arachidonic Acid 46-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 26975734-1 2016 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to generate epoxyeicosatrienoic acids (EETs). epoxyeicosatrienoic acids 75-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26975734-1 2016 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to generate epoxyeicosatrienoic acids (EETs). epoxyeicosatrienoic acids 75-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 26931777-11 2016 Based upon in vitro studies, the potential for LCZ696 to inhibit or induce cytochrome P450 (CYP) enzymes and cause CYP-mediated drug interactions clinically was found to be low. sacubitril and valsartan sodium hydrate drug combination 47-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 26975734-1 2016 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to generate epoxyeicosatrienoic acids (EETs). eets 102-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26931777-11 2016 Based upon in vitro studies, the potential for LCZ696 to inhibit or induce cytochrome P450 (CYP) enzymes and cause CYP-mediated drug interactions clinically was found to be low. sacubitril and valsartan sodium hydrate drug combination 47-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 26931777-11 2016 Based upon in vitro studies, the potential for LCZ696 to inhibit or induce cytochrome P450 (CYP) enzymes and cause CYP-mediated drug interactions clinically was found to be low. sacubitril and valsartan sodium hydrate drug combination 47-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 26975734-1 2016 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to generate epoxyeicosatrienoic acids (EETs). eets 102-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 26975734-3 2016 In addition to arachidonic acid, the CYP epoxygenases also metabolize the Omega-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid and docosahexaenoic acid, to generate bioactive lipid epoxide mediators. Arachidonic Acid 15-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 26975734-3 2016 In addition to arachidonic acid, the CYP epoxygenases also metabolize the Omega-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid and docosahexaenoic acid, to generate bioactive lipid epoxide mediators. omega-3 polyunsaturated fatty acids 74-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 26975734-3 2016 In addition to arachidonic acid, the CYP epoxygenases also metabolize the Omega-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid and docosahexaenoic acid, to generate bioactive lipid epoxide mediators. Fatty Acids, Unsaturated 111-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 26975734-3 2016 In addition to arachidonic acid, the CYP epoxygenases also metabolize the Omega-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid and docosahexaenoic acid, to generate bioactive lipid epoxide mediators. Eicosapentaenoic Acid 119-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 26975734-3 2016 In addition to arachidonic acid, the CYP epoxygenases also metabolize the Omega-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid and docosahexaenoic acid, to generate bioactive lipid epoxide mediators. Docosahexaenoic Acids 145-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 26975734-3 2016 In addition to arachidonic acid, the CYP epoxygenases also metabolize the Omega-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid and docosahexaenoic acid, to generate bioactive lipid epoxide mediators. lipid epoxide 189-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 26975734-6 2016 It seems that the overall consequences of CYP epoxygenase activation largely depend on enzyme substrate preference and the endogenous Omega-3/Omega-6 PUFA ratio. omega-3 134-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 26975734-6 2016 It seems that the overall consequences of CYP epoxygenase activation largely depend on enzyme substrate preference and the endogenous Omega-3/Omega-6 PUFA ratio. omega-6 142-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 27769286-6 2016 AGEs are associated with hyperandrogenism in PCOS possibly by altering the activity of various enzymes such as cholesterol side-chain cleavage enzyme cytochrome P450, steroidogenic acute regulatory protein, 17alpha-hydroxylase, and 3beta-hydroxysteroid dehydrogenase. Cholesterol 111-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-197 26961113-0 2016 Association of Cytochrome P450 Genetic Variants with Clopidogrel Resistance and Outcomes in Acute Ischemic Stroke. Clopidogrel 53-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-30 27474842-2 2016 Cytochrome P450 (CYP) enzymes convert losartan to E3174 active metabolite. Losartan 38-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26750984-4 2016 Although there have been studies on the biological effects of glycyrol, the inhibitory effects of glycyrol on cytochrome P450 (CYP) activities have not been investigated. glycyrol 98-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 26750984-4 2016 Although there have been studies on the biological effects of glycyrol, the inhibitory effects of glycyrol on cytochrome P450 (CYP) activities have not been investigated. glycyrol 98-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 26750984-6 2016 We investigated the inhibitory effects of glycyrol on the activities of CYP isoforms using a cocktail of probe substrates in pooled human liver microsome (HLM) and human recombinant cDNA-expressed CYPs. glycyrol 42-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 27009433-10 2016 In conclusion, Geneva cocktail combined with an innovative DBS sampling device can be used routinely as a test for simultaneous CYP phenotyping. dbs 59-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 27474842-2 2016 Cytochrome P450 (CYP) enzymes convert losartan to E3174 active metabolite. losartan carboxylic acid 50-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27458223-2 2016 Three studies were conducted to evaluate the potential drug-drug interactions between delamanid and antiretroviral drugs, including ritonavir, a strong inhibitor of CYP3A4, and selected anti-TB drugs, including rifampin, a strong inducer of cytochrome P450 (CYP) isozymes. OPC-67683 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-256 27458223-2 2016 Three studies were conducted to evaluate the potential drug-drug interactions between delamanid and antiretroviral drugs, including ritonavir, a strong inhibitor of CYP3A4, and selected anti-TB drugs, including rifampin, a strong inducer of cytochrome P450 (CYP) isozymes. OPC-67683 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-168 27458223-2 2016 Three studies were conducted to evaluate the potential drug-drug interactions between delamanid and antiretroviral drugs, including ritonavir, a strong inhibitor of CYP3A4, and selected anti-TB drugs, including rifampin, a strong inducer of cytochrome P450 (CYP) isozymes. Rifampin 211-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-256 27318274-0 2016 Flavokawain A inhibits Cytochrome P450 in in vitro metabolic and inhibitory investigations. flavokawain A 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 27318274-3 2016 AIM OF THE STUDY: The current study was purposed to investigate the interaction between Flavokawain A and Cytochrome P450, including the inhibitory effects of Flavokawain A on predominant CYP450 isotypes and further clarified the inhibitory mechanism of FKA on CYP450 enzymes. flavokawain A 88-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 27318274-3 2016 AIM OF THE STUDY: The current study was purposed to investigate the interaction between Flavokawain A and Cytochrome P450, including the inhibitory effects of Flavokawain A on predominant CYP450 isotypes and further clarified the inhibitory mechanism of FKA on CYP450 enzymes. BENZYL-CARBAMIC ACID [8-DEETHYL-ASCOMYCIN-8-YL]ETHYL ESTER 254-257 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 27323294-1 2016 Cytochrome P450 (P450) 3A (CYP3A) is an enzyme responsible for the metabolism of therapeutic drugs such as midazolam, nifedipine, testosterone and triazolam. Midazolam 107-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-25 27323294-1 2016 Cytochrome P450 (P450) 3A (CYP3A) is an enzyme responsible for the metabolism of therapeutic drugs such as midazolam, nifedipine, testosterone and triazolam. Nifedipine 118-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-25 27323294-1 2016 Cytochrome P450 (P450) 3A (CYP3A) is an enzyme responsible for the metabolism of therapeutic drugs such as midazolam, nifedipine, testosterone and triazolam. Testosterone 130-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-25 27323294-1 2016 Cytochrome P450 (P450) 3A (CYP3A) is an enzyme responsible for the metabolism of therapeutic drugs such as midazolam, nifedipine, testosterone and triazolam. Triazolam 147-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-25 27474842-2 2016 Cytochrome P450 (CYP) enzymes convert losartan to E3174 active metabolite. losartan carboxylic acid 50-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 27474842-6 2016 Differences in tolerated losartan daily doses with respect to CYP metabolic classes were assessed through the Kruskal-Wallis test. Losartan 25-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 27474842-2 2016 Cytochrome P450 (CYP) enzymes convert losartan to E3174 active metabolite. Losartan 38-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 27101231-1 2016 Objectives: To determine relations between drug concentrations and the cytochrome P450-CYP2D6 genotype or phenotype among elderly patients treated with nortriptyline or venlafaxine. Nortriptyline 152-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-93 27171763-8 2016 Our results suggest that caution may be warranted when CYP-related co-medications are administered during sildenafil treatment for PH. Sildenafil Citrate 106-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 27795624-2 2016 Rifampicin, a first-line antitubercular therapy (ATT) drug, is a potent inducer of hepatic cytochrome P450 (CYP). Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 27795624-2 2016 Rifampicin, a first-line antitubercular therapy (ATT) drug, is a potent inducer of hepatic cytochrome P450 (CYP). Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 27795624-3 2016 There is potential for pharmacokinetic interaction between rifampicin and anti-hypertensives that are CYP substrates: amlodipine and metoprolol. Rifampin 59-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 27795624-3 2016 There is potential for pharmacokinetic interaction between rifampicin and anti-hypertensives that are CYP substrates: amlodipine and metoprolol. Amlodipine 118-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 27795624-3 2016 There is potential for pharmacokinetic interaction between rifampicin and anti-hypertensives that are CYP substrates: amlodipine and metoprolol. Metoprolol 133-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 27101231-1 2016 Objectives: To determine relations between drug concentrations and the cytochrome P450-CYP2D6 genotype or phenotype among elderly patients treated with nortriptyline or venlafaxine. Venlafaxine Hydrochloride 169-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-93 27251443-1 2016 Cytochrome P450 (CYP) enzymes play important roles in the metabolism of exogenous compounds such as polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 100-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27387538-5 2016 Of the CYP enzymes tested, CYP2B6-catalyzed bupropion 6-hydroxylation was inhibited by S002-333 (IC50 ~ 9.25 +- 2.46 muM) in a stereoselective manner with (S)-isomer showing potent inhibition (IC50 ~ 5.28 +- 1.25 muM) in contrast to (R)-isomer which showed negligible inhibition on CYP2B6 activity (IC50 > 50 muM). Bupropion 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-10 27251443-1 2016 Cytochrome P450 (CYP) enzymes play important roles in the metabolism of exogenous compounds such as polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 100-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 27251443-1 2016 Cytochrome P450 (CYP) enzymes play important roles in the metabolism of exogenous compounds such as polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 134-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27251443-1 2016 Cytochrome P450 (CYP) enzymes play important roles in the metabolism of exogenous compounds such as polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 134-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 26360322-0 2016 Cytochrome P450-mediated metabolism of the synthetic cannabinoids UR-144 and XLR-11. (1-pentyl-1H-indol-3-yl)(2,2,3,3-tetramethylcyclopropyl)methanone 66-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27516570-13 2016 Apart from hydrolytic degradation, endocannabinoids may also be oxidized by cyclooxygenase-2, lipoxygenases, and cytochrome P450. Endocannabinoids 35-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 26360322-4 2016 This study extends previous studies by identifying the specific CYP enzymes involved in the metabolism of UR-144 and XLR-11 utilizing a panel of nine recombinant enzymes (CYP1A2, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 3A4, and 2E1). Urea 106-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 26360322-6 2016 Incubations of UR-144 and XLR-11 with recombinant CYP enzymes revealed that UR-144 and XLR-11 are extensively metabolized by CYP3A4 at the tetramethylcyclopropyl (TMCP) moiety, but also CYP1A2 and CYP2C19 showed activity. Urea 15-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 26360322-6 2016 Incubations of UR-144 and XLR-11 with recombinant CYP enzymes revealed that UR-144 and XLR-11 are extensively metabolized by CYP3A4 at the tetramethylcyclopropyl (TMCP) moiety, but also CYP1A2 and CYP2C19 showed activity. Urea 76-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 27179126-9 2016 Dasabuvir was cleared mainly through cytochrome P450-mediated oxidation metabolism to M1. dasabuvir 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 27179126-12 2016 Cytochrome P450 profiling indicated that dasabuvir was mainly metabolized by CYP2C8, followed by CYP3A4. dasabuvir 41-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26360322-6 2016 Incubations of UR-144 and XLR-11 with recombinant CYP enzymes revealed that UR-144 and XLR-11 are extensively metabolized by CYP3A4 at the tetramethylcyclopropyl (TMCP) moiety, but also CYP1A2 and CYP2C19 showed activity. tmcp 163-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 26360322-7 2016 Inhibition of CYP3A4 in HLM attenuated the metabolism of UR-144 and XLR-11, while inhibition of the other CYP enzymes in HLM had only minor effects. Urea 57-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 26360322-8 2016 Thus, CYP3A4 is the major contributor to the CYP mediated metabolism of UR-144 and XLR-11 with minor contributions from CYP1A2. Urea 72-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Benzodiazepines 96-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27286171-1 2016 Polyunsaturated fatty acids (PUFAs) undergo cytochrome P450 (CYP)-dependent oxidation to epoxides that modulate important physiological functions, including vasoactivity, inflammation, nociception, proliferation and viability. Fatty Acids, Unsaturated 0-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 27286171-1 2016 Polyunsaturated fatty acids (PUFAs) undergo cytochrome P450 (CYP)-dependent oxidation to epoxides that modulate important physiological functions, including vasoactivity, inflammation, nociception, proliferation and viability. Fatty Acids, Unsaturated 0-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 27286171-1 2016 Polyunsaturated fatty acids (PUFAs) undergo cytochrome P450 (CYP)-dependent oxidation to epoxides that modulate important physiological functions, including vasoactivity, inflammation, nociception, proliferation and viability. Epoxy Compounds 89-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 27286171-1 2016 Polyunsaturated fatty acids (PUFAs) undergo cytochrome P450 (CYP)-dependent oxidation to epoxides that modulate important physiological functions, including vasoactivity, inflammation, nociception, proliferation and viability. Epoxy Compounds 89-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Benzodiazepines 96-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Benzodiazepines 113-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Benzodiazepines 113-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Carbamazepine 120-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Carbamazepine 120-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Carbamazepine 135-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Carbamazepine 135-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Phenytoin 141-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Phenytoin 141-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Phenytoin 152-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Phenytoin 152-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Phenobarbital 157-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Phenobarbital 157-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Phenobarbital 172-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Phenobarbital 172-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Valproic Acid 181-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Valproic Acid 181-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Valproic Acid 196-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27490534-3 2016 Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). Valproic Acid 196-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27490534-4 2016 The induction of CYP450 isoenzymes may cause vitamin D deficiency, hypocalcemia, increased fracture risks, and altered bone turnover, leading to impaired bone mineral density (BMD). Vitamin D 45-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 27267697-1 2016 Cytochrome P450 (P450) enzymes are important in the metabolism of drugs, steroids, fat-soluble vitamins, carcinogens, pesticides, and many other types of chemicals. Steroids 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27467145-1 2016 Although Plasmodium vivax relapses are classically associated with hypnozoite activation, it has been proposed that a proportion of these cases are due to primaquine (PQ) treatment failure caused by polymorphisms in cytochrome P-450 2D6 (CYP2D6). Primaquine 155-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-232 27467145-1 2016 Although Plasmodium vivax relapses are classically associated with hypnozoite activation, it has been proposed that a proportion of these cases are due to primaquine (PQ) treatment failure caused by polymorphisms in cytochrome P-450 2D6 (CYP2D6). Primaquine 167-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-232 26945713-3 2016 Cobicistat and ritonavir are equally strong inhibitors of cytochrome P450 (CYP) 3A4 and consequently were shown to be equivalent pharmacokinetic enhancers for elvitegravir and for the PIs atazanavir and darunavir. Cobicistat 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 27347790-0 2016 Effect of Cholesterol on the Interaction of Cytochrome P450 Substrate Drug Chlorzoxazone with the Phosphatidylcholine Bilayer. Cholesterol 10-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 27347790-0 2016 Effect of Cholesterol on the Interaction of Cytochrome P450 Substrate Drug Chlorzoxazone with the Phosphatidylcholine Bilayer. Chlorzoxazone 75-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 27347790-0 2016 Effect of Cholesterol on the Interaction of Cytochrome P450 Substrate Drug Chlorzoxazone with the Phosphatidylcholine Bilayer. Phosphatidylcholines 98-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 27347790-6 2016 In this study, the cholesterol concentration dependence of the interaction of a CYP substrate drug, chlorzoxazone (CZX), with model membranes composed of phosphatidylcholine (PC) and cholesterol was examined via differential scanning calorimetry (DSC), UV-visible spectroscopy, and X-ray diffraction. Cholesterol 19-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 27347790-6 2016 In this study, the cholesterol concentration dependence of the interaction of a CYP substrate drug, chlorzoxazone (CZX), with model membranes composed of phosphatidylcholine (PC) and cholesterol was examined via differential scanning calorimetry (DSC), UV-visible spectroscopy, and X-ray diffraction. Chlorzoxazone 100-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 27347790-6 2016 In this study, the cholesterol concentration dependence of the interaction of a CYP substrate drug, chlorzoxazone (CZX), with model membranes composed of phosphatidylcholine (PC) and cholesterol was examined via differential scanning calorimetry (DSC), UV-visible spectroscopy, and X-ray diffraction. Chlorzoxazone 115-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 27347790-6 2016 In this study, the cholesterol concentration dependence of the interaction of a CYP substrate drug, chlorzoxazone (CZX), with model membranes composed of phosphatidylcholine (PC) and cholesterol was examined via differential scanning calorimetry (DSC), UV-visible spectroscopy, and X-ray diffraction. Phosphatidylcholines 154-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 27347790-6 2016 In this study, the cholesterol concentration dependence of the interaction of a CYP substrate drug, chlorzoxazone (CZX), with model membranes composed of phosphatidylcholine (PC) and cholesterol was examined via differential scanning calorimetry (DSC), UV-visible spectroscopy, and X-ray diffraction. Phosphatidylcholines 175-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 27347790-6 2016 In this study, the cholesterol concentration dependence of the interaction of a CYP substrate drug, chlorzoxazone (CZX), with model membranes composed of phosphatidylcholine (PC) and cholesterol was examined via differential scanning calorimetry (DSC), UV-visible spectroscopy, and X-ray diffraction. Cholesterol 183-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 27347790-11 2016 The low cholesterol contents (12-20 mol %) of ER membranes might be the most suitable for the CYP drug metabolism process in ER membranes. Cholesterol 8-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 27185541-1 2016 The purpose of this work was to investigate the in vitro metabolism of nitracaine, a new psychoactive substance, using human liver microsome incubations, to evaluate the cytochrome P450 (CYP) enzyme isoforms responsible for the phase-I metabolism and to compare the information from the in vitro experiments with data resulting from an authentic user"s urine sample. Nitracaine 71-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-185 27185541-1 2016 The purpose of this work was to investigate the in vitro metabolism of nitracaine, a new psychoactive substance, using human liver microsome incubations, to evaluate the cytochrome P450 (CYP) enzyme isoforms responsible for the phase-I metabolism and to compare the information from the in vitro experiments with data resulting from an authentic user"s urine sample. Nitracaine 71-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 187-190 26945713-3 2016 Cobicistat and ritonavir are equally strong inhibitors of cytochrome P450 (CYP) 3A4 and consequently were shown to be equivalent pharmacokinetic enhancers for elvitegravir and for the PIs atazanavir and darunavir. Cobicistat 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 26945713-3 2016 Cobicistat and ritonavir are equally strong inhibitors of cytochrome P450 (CYP) 3A4 and consequently were shown to be equivalent pharmacokinetic enhancers for elvitegravir and for the PIs atazanavir and darunavir. Ritonavir 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 26945713-3 2016 Cobicistat and ritonavir are equally strong inhibitors of cytochrome P450 (CYP) 3A4 and consequently were shown to be equivalent pharmacokinetic enhancers for elvitegravir and for the PIs atazanavir and darunavir. Ritonavir 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 26945713-4 2016 Since cobicistat is a more selective CYP inhibitor than ritonavir and is devoid of enzyme-inducing properties, differences are expected in their interaction profiles with some co-medications. Cobicistat 6-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 27212636-4 2016 The total cytochrome P450 activity was determined by measuring the transformation of 7-ethoxycoumarin. 7-ethoxycoumarin 85-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 27115337-1 2016 BACKGROUND: Epoxyeicosatrienoic acids (EETs) derived from cytochrome P450 (CYP)-dependent metabolism of arachidonic acid are increased in the plasma of women with preeclampsia as compared with normal pregnancy and are significantly higher in fetal than in maternal plasma and erythrocytes. epoxyeicosatrienoic acids 12-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 27115337-1 2016 BACKGROUND: Epoxyeicosatrienoic acids (EETs) derived from cytochrome P450 (CYP)-dependent metabolism of arachidonic acid are increased in the plasma of women with preeclampsia as compared with normal pregnancy and are significantly higher in fetal than in maternal plasma and erythrocytes. epoxyeicosatrienoic acids 12-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 27115337-1 2016 BACKGROUND: Epoxyeicosatrienoic acids (EETs) derived from cytochrome P450 (CYP)-dependent metabolism of arachidonic acid are increased in the plasma of women with preeclampsia as compared with normal pregnancy and are significantly higher in fetal than in maternal plasma and erythrocytes. eets 39-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 27115337-1 2016 BACKGROUND: Epoxyeicosatrienoic acids (EETs) derived from cytochrome P450 (CYP)-dependent metabolism of arachidonic acid are increased in the plasma of women with preeclampsia as compared with normal pregnancy and are significantly higher in fetal than in maternal plasma and erythrocytes. eets 39-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 27115337-1 2016 BACKGROUND: Epoxyeicosatrienoic acids (EETs) derived from cytochrome P450 (CYP)-dependent metabolism of arachidonic acid are increased in the plasma of women with preeclampsia as compared with normal pregnancy and are significantly higher in fetal than in maternal plasma and erythrocytes. Arachidonic Acid 104-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 27115337-1 2016 BACKGROUND: Epoxyeicosatrienoic acids (EETs) derived from cytochrome P450 (CYP)-dependent metabolism of arachidonic acid are increased in the plasma of women with preeclampsia as compared with normal pregnancy and are significantly higher in fetal than in maternal plasma and erythrocytes. Arachidonic Acid 104-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 27113703-2 2016 To investigate the role of drug metabolism in this liver toxicity, amiodarone and its major metabolite desethylamiodarone were incubated with HepG2 cells overexpressing a series of cytochrome P450 (CYP) isoforms. desethylamiodarone 103-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-201 26965288-8 2016 Leukotoxin levels correlated with goblet cell abundance (r=0.72; p=0.028).These findings suggest a mechanism by which goblet cell-associated cytochrome-P450 and sEH activity produce elevated leukotoxin-diol levels, which play a putative role in the clinical manifestations of COPD in a female-dominated disease sub-phenotype. diol 202-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-156 27060436-2 2016 Rasagiline is metabolized by the cytochrome P450 (CYP) system, and the following three major metabolites with potential neuroprotective activity have been identified: 1-aminoindan (AI), 3-hydroxy-N-propargyl-1-aminoindan (3-OH-PAI) and 3-hydroxy-1-aminoindan (3-OH-AI). rasagiline 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 27060436-2 2016 Rasagiline is metabolized by the cytochrome P450 (CYP) system, and the following three major metabolites with potential neuroprotective activity have been identified: 1-aminoindan (AI), 3-hydroxy-N-propargyl-1-aminoindan (3-OH-PAI) and 3-hydroxy-1-aminoindan (3-OH-AI). rasagiline 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 27029743-4 2016 Cytochrome P450s were the main enzymes involved in the metabolism of daclatasvir. daclatasvir 69-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26921441-1 2016 20-Hydroxyeicosatetraeonic acid (20-HETE) produced by cytochrome P-450 monooxygenases in NADPH-dependent manner is proinflammatory, and it contributes to the pathogenesis of systemic and pulmonary hypertension. 20-Hete 0-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 27100515-1 2016 Cytochrome P450s enzymes catalyze the metabolism of arachidonic acid to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid and hydroxyeicosatetraeonic acid (HETEs). Arachidonic Acid 52-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27100515-1 2016 Cytochrome P450s enzymes catalyze the metabolism of arachidonic acid to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid and hydroxyeicosatetraeonic acid (HETEs). epoxyeicosatrienoic acids 72-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27100515-1 2016 Cytochrome P450s enzymes catalyze the metabolism of arachidonic acid to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid and hydroxyeicosatetraeonic acid (HETEs). eets 99-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27100515-1 2016 Cytochrome P450s enzymes catalyze the metabolism of arachidonic acid to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid and hydroxyeicosatetraeonic acid (HETEs). dihydroxyeicosatetraenoic acid 106-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27100515-1 2016 Cytochrome P450s enzymes catalyze the metabolism of arachidonic acid to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid and hydroxyeicosatetraeonic acid (HETEs). hydroxyeicosatetraeonic acid 141-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27100515-1 2016 Cytochrome P450s enzymes catalyze the metabolism of arachidonic acid to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid and hydroxyeicosatetraeonic acid (HETEs). Hydroxyeicosatetraenoic Acids 171-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27063220-5 2016 Direct CYP inhibition was validated using 7 inhibitors (alpha-naphthoflavone, tranylcypromine, ticlopidine, fluconazole, quinidine, ketoconazole and 1-ABT). alpha-naphthoflavone 56-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-10 27063220-5 2016 Direct CYP inhibition was validated using 7 inhibitors (alpha-naphthoflavone, tranylcypromine, ticlopidine, fluconazole, quinidine, ketoconazole and 1-ABT). Tranylcypromine 78-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-10 27063220-5 2016 Direct CYP inhibition was validated using 7 inhibitors (alpha-naphthoflavone, tranylcypromine, ticlopidine, fluconazole, quinidine, ketoconazole and 1-ABT). Ticlopidine 95-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-10 27063220-5 2016 Direct CYP inhibition was validated using 7 inhibitors (alpha-naphthoflavone, tranylcypromine, ticlopidine, fluconazole, quinidine, ketoconazole and 1-ABT). Fluconazole 108-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-10 27063220-5 2016 Direct CYP inhibition was validated using 7 inhibitors (alpha-naphthoflavone, tranylcypromine, ticlopidine, fluconazole, quinidine, ketoconazole and 1-ABT). Quinidine 121-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-10 27063220-5 2016 Direct CYP inhibition was validated using 7 inhibitors (alpha-naphthoflavone, tranylcypromine, ticlopidine, fluconazole, quinidine, ketoconazole and 1-ABT). Ketoconazole 132-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-10 27063220-5 2016 Direct CYP inhibition was validated using 7 inhibitors (alpha-naphthoflavone, tranylcypromine, ticlopidine, fluconazole, quinidine, ketoconazole and 1-ABT). 1-abt 149-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-10 26921441-1 2016 20-Hydroxyeicosatetraeonic acid (20-HETE) produced by cytochrome P-450 monooxygenases in NADPH-dependent manner is proinflammatory, and it contributes to the pathogenesis of systemic and pulmonary hypertension. 20-Hete 33-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 26921441-1 2016 20-Hydroxyeicosatetraeonic acid (20-HETE) produced by cytochrome P-450 monooxygenases in NADPH-dependent manner is proinflammatory, and it contributes to the pathogenesis of systemic and pulmonary hypertension. NADP 89-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 26921388-1 2016 Replacement of hydrogen with fluorine is a useful drug design strategy when decreases in cytochrome P450 (P450) metabolic lability are needed. Hydrogen 15-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 27265124-0 2016 Maintenance of High Cytochrome P450 Expression in HepaRG Cell Spheroids in DMSO-Free Medium. Dimethyl Sulfoxide 75-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 26921388-1 2016 Replacement of hydrogen with fluorine is a useful drug design strategy when decreases in cytochrome P450 (P450) metabolic lability are needed. Fluorine 29-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 27079253-3 2016 Epoxyecosatrienoic acids (EETs), the products of arachidonic acid metabolism mediated by cytochrome P450 (CYP) 2J, 2C and other isoforms, are regulated by soluble epoxide hydrolase (sEH)-catalyzed conversion into less active diols. epoxyecosatrienoic acids 0-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 27079253-3 2016 Epoxyecosatrienoic acids (EETs), the products of arachidonic acid metabolism mediated by cytochrome P450 (CYP) 2J, 2C and other isoforms, are regulated by soluble epoxide hydrolase (sEH)-catalyzed conversion into less active diols. epoxyecosatrienoic acids 0-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 27079253-3 2016 Epoxyecosatrienoic acids (EETs), the products of arachidonic acid metabolism mediated by cytochrome P450 (CYP) 2J, 2C and other isoforms, are regulated by soluble epoxide hydrolase (sEH)-catalyzed conversion into less active diols. eets 26-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 27079253-3 2016 Epoxyecosatrienoic acids (EETs), the products of arachidonic acid metabolism mediated by cytochrome P450 (CYP) 2J, 2C and other isoforms, are regulated by soluble epoxide hydrolase (sEH)-catalyzed conversion into less active diols. eets 26-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 27079253-3 2016 Epoxyecosatrienoic acids (EETs), the products of arachidonic acid metabolism mediated by cytochrome P450 (CYP) 2J, 2C and other isoforms, are regulated by soluble epoxide hydrolase (sEH)-catalyzed conversion into less active diols. Arachidonic Acid 49-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 27079253-3 2016 Epoxyecosatrienoic acids (EETs), the products of arachidonic acid metabolism mediated by cytochrome P450 (CYP) 2J, 2C and other isoforms, are regulated by soluble epoxide hydrolase (sEH)-catalyzed conversion into less active diols. Arachidonic Acid 49-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 27079253-3 2016 Epoxyecosatrienoic acids (EETs), the products of arachidonic acid metabolism mediated by cytochrome P450 (CYP) 2J, 2C and other isoforms, are regulated by soluble epoxide hydrolase (sEH)-catalyzed conversion into less active diols. diols 225-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 27079253-3 2016 Epoxyecosatrienoic acids (EETs), the products of arachidonic acid metabolism mediated by cytochrome P450 (CYP) 2J, 2C and other isoforms, are regulated by soluble epoxide hydrolase (sEH)-catalyzed conversion into less active diols. diols 225-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 26714750-7 2016 The activation of cytochrome P450 (CYP) enzymes in HepG2 cells grown on CBD-CAS-coated honeycomb collagen sponge was measured at the mRNA level and was found to increase between two and six times compared to cells grown without the CBD-CAS coating, showing that this culture system induced CYP gene expression and thus may be useful in drug metabolism assays. cbd-cas 72-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 26969934-2 2016 It is bioactivated by cytochrome P450 (CYP) enzymes to reactive metabolites, which may further react with glutathione to form S-linked and N-linked conjugates. Glutathione 106-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 26969934-2 2016 It is bioactivated by cytochrome P450 (CYP) enzymes to reactive metabolites, which may further react with glutathione to form S-linked and N-linked conjugates. Glutathione 106-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 26969934-6 2016 Menthofuran was also incubated with recombinant human CYP enzymes and GSH to elucidate the CYPs responsible for the formation of the reactive metabolites. menthofuran 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 26969934-9 2016 CYP1A2, 2B6 and 3A4 were observed to produce more GSH conjugates than other CYP isoforms. Glutathione 50-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 27128896-0 2016 Inhibitory Effects of Aschantin on Cytochrome P450 and Uridine 5"-diphospho-glucuronosyltransferase Enzyme Activities in Human Liver Microsomes. aschantin 22-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 26896706-2 2016 TAM is metabolized to the more active 4-hydroxytamoxifen (4-OH-TAM) and endoxifen by cytochrome P450 (CYP) mainly CYP2D6 and CYP3A4 enzymes. Tamoxifen 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 26896706-2 2016 TAM is metabolized to the more active 4-hydroxytamoxifen (4-OH-TAM) and endoxifen by cytochrome P450 (CYP) mainly CYP2D6 and CYP3A4 enzymes. Tamoxifen 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 26896706-2 2016 TAM is metabolized to the more active 4-hydroxytamoxifen (4-OH-TAM) and endoxifen by cytochrome P450 (CYP) mainly CYP2D6 and CYP3A4 enzymes. hydroxytamoxifen 38-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 26896706-2 2016 TAM is metabolized to the more active 4-hydroxytamoxifen (4-OH-TAM) and endoxifen by cytochrome P450 (CYP) mainly CYP2D6 and CYP3A4 enzymes. hydroxytamoxifen 38-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 26896706-2 2016 TAM is metabolized to the more active 4-hydroxytamoxifen (4-OH-TAM) and endoxifen by cytochrome P450 (CYP) mainly CYP2D6 and CYP3A4 enzymes. 4-hydroxy-N-desmethyltamoxifen 72-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 26896706-2 2016 TAM is metabolized to the more active 4-hydroxytamoxifen (4-OH-TAM) and endoxifen by cytochrome P450 (CYP) mainly CYP2D6 and CYP3A4 enzymes. 4-hydroxy-N-desmethyltamoxifen 72-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 26690878-0 2016 Mutating a Highly Conserved Residue in Diverse Cytochrome P450s Facilitates Diastereoselective Olefin Cyclopropanation. Alkenes 95-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 26714750-7 2016 The activation of cytochrome P450 (CYP) enzymes in HepG2 cells grown on CBD-CAS-coated honeycomb collagen sponge was measured at the mRNA level and was found to increase between two and six times compared to cells grown without the CBD-CAS coating, showing that this culture system induced CYP gene expression and thus may be useful in drug metabolism assays. cbd-cas 72-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 26714750-7 2016 The activation of cytochrome P450 (CYP) enzymes in HepG2 cells grown on CBD-CAS-coated honeycomb collagen sponge was measured at the mRNA level and was found to increase between two and six times compared to cells grown without the CBD-CAS coating, showing that this culture system induced CYP gene expression and thus may be useful in drug metabolism assays. cbd-cas 72-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 290-293 26714750-7 2016 The activation of cytochrome P450 (CYP) enzymes in HepG2 cells grown on CBD-CAS-coated honeycomb collagen sponge was measured at the mRNA level and was found to increase between two and six times compared to cells grown without the CBD-CAS coating, showing that this culture system induced CYP gene expression and thus may be useful in drug metabolism assays. cbd-cas 232-239 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 26714750-7 2016 The activation of cytochrome P450 (CYP) enzymes in HepG2 cells grown on CBD-CAS-coated honeycomb collagen sponge was measured at the mRNA level and was found to increase between two and six times compared to cells grown without the CBD-CAS coating, showing that this culture system induced CYP gene expression and thus may be useful in drug metabolism assays. cbd-cas 232-239 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 26753974-3 2016 Metabolic efficiency of the mutant library towards a selection of CYP model substrates, being amitriptyline (AMI), buspirone (BUS), coumarine (COU), dextromethorphan (DEX), diclofenac (DIC) and norethisterone (NET), was investigated. Amitriptyline 94-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 26633836-4 2016 However, VKAs can interfere with clopidogrel metabolism via the cytochrome P450 (CYP) system which in turn may result in an increase in platelet reactivity. Clopidogrel 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 26633836-4 2016 However, VKAs can interfere with clopidogrel metabolism via the cytochrome P450 (CYP) system which in turn may result in an increase in platelet reactivity. Clopidogrel 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 26316040-11 2016 CYP polymorphisms may explain why there is wide variation in blood HCQ concentrations. Hydroxychloroquine 67-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26658226-0 2016 Metabolism of Oral Turinabol by Human Steroid Hormone-Synthesizing Cytochrome P450 Enzymes. turinabol 19-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 26658226-0 2016 Metabolism of Oral Turinabol by Human Steroid Hormone-Synthesizing Cytochrome P450 Enzymes. Steroids 38-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 26658226-1 2016 The human mitochondrial cytochrome P450 enzymes CYP11A1, CYP11B1, and CYP11B2 are involved in the biosynthesis of steroid hormones. Steroids 114-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 26727525-0 2016 Chronic Effects of Ethanol and/or Darunavir/Ritonavir on U937 Monocytic Cells: Regulation of Cytochrome P450 and Antioxidant Enzymes, Oxidative Stress, and Cytotoxicity. Ethanol 19-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-108 26727525-0 2016 Chronic Effects of Ethanol and/or Darunavir/Ritonavir on U937 Monocytic Cells: Regulation of Cytochrome P450 and Antioxidant Enzymes, Oxidative Stress, and Cytotoxicity. Darunavir 34-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-108 26727525-4 2016 METHODS: This study examined the chronic effects of EtOH and ART (darunavir/ritonavir), alone and in combination, on expression/levels of cytochrome P450 enzymes (CYPs), antioxidant enzymes (AOEs), reactive oxygen species (ROS), and cytotoxicity in U937 cells. Ethanol 52-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-153 26727525-4 2016 METHODS: This study examined the chronic effects of EtOH and ART (darunavir/ritonavir), alone and in combination, on expression/levels of cytochrome P450 enzymes (CYPs), antioxidant enzymes (AOEs), reactive oxygen species (ROS), and cytotoxicity in U937 cells. art 61-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-153 26727525-4 2016 METHODS: This study examined the chronic effects of EtOH and ART (darunavir/ritonavir), alone and in combination, on expression/levels of cytochrome P450 enzymes (CYPs), antioxidant enzymes (AOEs), reactive oxygen species (ROS), and cytotoxicity in U937 cells. Darunavir 66-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-153 26727525-4 2016 METHODS: This study examined the chronic effects of EtOH and ART (darunavir/ritonavir), alone and in combination, on expression/levels of cytochrome P450 enzymes (CYPs), antioxidant enzymes (AOEs), reactive oxygen species (ROS), and cytotoxicity in U937 cells. Ritonavir 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-153 26316040-1 2016 OBJECTIVE: To evaluate associations of genetic polymorphisms in cytochrome P450 (CYP) isoforms 2D6, 3A5, and 3A4 with blood concentrations of hydroxychloroquine (HCQ) and its metabolite, N-desethyl HCQ (DHCQ), in patients with systemic lupus erythematosus (SLE). Hydroxychloroquine 142-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 26316040-1 2016 OBJECTIVE: To evaluate associations of genetic polymorphisms in cytochrome P450 (CYP) isoforms 2D6, 3A5, and 3A4 with blood concentrations of hydroxychloroquine (HCQ) and its metabolite, N-desethyl HCQ (DHCQ), in patients with systemic lupus erythematosus (SLE). Hydroxychloroquine 142-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 26316040-1 2016 OBJECTIVE: To evaluate associations of genetic polymorphisms in cytochrome P450 (CYP) isoforms 2D6, 3A5, and 3A4 with blood concentrations of hydroxychloroquine (HCQ) and its metabolite, N-desethyl HCQ (DHCQ), in patients with systemic lupus erythematosus (SLE). Hydroxychloroquine 162-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 26316040-1 2016 OBJECTIVE: To evaluate associations of genetic polymorphisms in cytochrome P450 (CYP) isoforms 2D6, 3A5, and 3A4 with blood concentrations of hydroxychloroquine (HCQ) and its metabolite, N-desethyl HCQ (DHCQ), in patients with systemic lupus erythematosus (SLE). Hydroxychloroquine 162-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 25398514-2 2016 We have investigated the role of TP53 in cytochrome P450 (CYP)-mediated metabolic activation of three polycyclic aromatic hydrocarbons (PAHs) in a panel of isogenic colorectal HCT116 cells with differing TP53 status. Polycyclic Aromatic Hydrocarbons 102-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 25398514-2 2016 We have investigated the role of TP53 in cytochrome P450 (CYP)-mediated metabolic activation of three polycyclic aromatic hydrocarbons (PAHs) in a panel of isogenic colorectal HCT116 cells with differing TP53 status. Polycyclic Aromatic Hydrocarbons 102-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 25398514-2 2016 We have investigated the role of TP53 in cytochrome P450 (CYP)-mediated metabolic activation of three polycyclic aromatic hydrocarbons (PAHs) in a panel of isogenic colorectal HCT116 cells with differing TP53 status. Polycyclic Aromatic Hydrocarbons 136-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 25398514-2 2016 We have investigated the role of TP53 in cytochrome P450 (CYP)-mediated metabolic activation of three polycyclic aromatic hydrocarbons (PAHs) in a panel of isogenic colorectal HCT116 cells with differing TP53 status. Polycyclic Aromatic Hydrocarbons 136-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 26446141-1 2016 BACKGROUND AND OBJECTIVE: The anti-oestrogen tamoxifen requires metabolic activation to endoxifen by cytochrome P450 (CYP) enzymes, predominantly CYP2D6. Tamoxifen 45-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 26446141-1 2016 BACKGROUND AND OBJECTIVE: The anti-oestrogen tamoxifen requires metabolic activation to endoxifen by cytochrome P450 (CYP) enzymes, predominantly CYP2D6. Tamoxifen 45-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 26446141-1 2016 BACKGROUND AND OBJECTIVE: The anti-oestrogen tamoxifen requires metabolic activation to endoxifen by cytochrome P450 (CYP) enzymes, predominantly CYP2D6. 4-hydroxy-N-desmethyltamoxifen 88-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 26446141-1 2016 BACKGROUND AND OBJECTIVE: The anti-oestrogen tamoxifen requires metabolic activation to endoxifen by cytochrome P450 (CYP) enzymes, predominantly CYP2D6. 4-hydroxy-N-desmethyltamoxifen 88-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 26820384-0 2016 Structure of OxyA tei: completing our picture of the glycopeptide antibiotic producing Cytochrome P450 cascade. Glycopeptides 53-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 26820384-1 2016 Cyclization of glycopeptide antibiotic precursors occurs in either three or four steps catalyzed by Cytochrome P450 enzymes. Glycopeptides 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 26316040-1 2016 OBJECTIVE: To evaluate associations of genetic polymorphisms in cytochrome P450 (CYP) isoforms 2D6, 3A5, and 3A4 with blood concentrations of hydroxychloroquine (HCQ) and its metabolite, N-desethyl HCQ (DHCQ), in patients with systemic lupus erythematosus (SLE). n-desethyl hcq 187-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 26316040-12 2016 The role of an individual"s CYP polymorphisms should be considered when prescribing oral HCQ. Hydroxychloroquine 89-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 26316040-1 2016 OBJECTIVE: To evaluate associations of genetic polymorphisms in cytochrome P450 (CYP) isoforms 2D6, 3A5, and 3A4 with blood concentrations of hydroxychloroquine (HCQ) and its metabolite, N-desethyl HCQ (DHCQ), in patients with systemic lupus erythematosus (SLE). n-desethyl hcq 187-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 26316040-1 2016 OBJECTIVE: To evaluate associations of genetic polymorphisms in cytochrome P450 (CYP) isoforms 2D6, 3A5, and 3A4 with blood concentrations of hydroxychloroquine (HCQ) and its metabolite, N-desethyl HCQ (DHCQ), in patients with systemic lupus erythematosus (SLE). dhcq 203-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 26316040-1 2016 OBJECTIVE: To evaluate associations of genetic polymorphisms in cytochrome P450 (CYP) isoforms 2D6, 3A5, and 3A4 with blood concentrations of hydroxychloroquine (HCQ) and its metabolite, N-desethyl HCQ (DHCQ), in patients with systemic lupus erythematosus (SLE). dhcq 203-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 27144772-1 2016 Soluble epoxide hydrolase (sEH) converts highly active epoxyeicosatrienoic acids (EETs) generated by cytochrome P450 (CYP) epoxygenases from arachidonic acid to less active dihydroxyeicosatrienoic acids. epoxyeicosatrienoic acids 55-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 27144772-1 2016 Soluble epoxide hydrolase (sEH) converts highly active epoxyeicosatrienoic acids (EETs) generated by cytochrome P450 (CYP) epoxygenases from arachidonic acid to less active dihydroxyeicosatrienoic acids. epoxyeicosatrienoic acids 55-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 27144772-1 2016 Soluble epoxide hydrolase (sEH) converts highly active epoxyeicosatrienoic acids (EETs) generated by cytochrome P450 (CYP) epoxygenases from arachidonic acid to less active dihydroxyeicosatrienoic acids. eets 82-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 27144772-1 2016 Soluble epoxide hydrolase (sEH) converts highly active epoxyeicosatrienoic acids (EETs) generated by cytochrome P450 (CYP) epoxygenases from arachidonic acid to less active dihydroxyeicosatrienoic acids. eets 82-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 27144772-1 2016 Soluble epoxide hydrolase (sEH) converts highly active epoxyeicosatrienoic acids (EETs) generated by cytochrome P450 (CYP) epoxygenases from arachidonic acid to less active dihydroxyeicosatrienoic acids. Arachidonic Acid 141-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 27144772-1 2016 Soluble epoxide hydrolase (sEH) converts highly active epoxyeicosatrienoic acids (EETs) generated by cytochrome P450 (CYP) epoxygenases from arachidonic acid to less active dihydroxyeicosatrienoic acids. Arachidonic Acid 141-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 27144772-1 2016 Soluble epoxide hydrolase (sEH) converts highly active epoxyeicosatrienoic acids (EETs) generated by cytochrome P450 (CYP) epoxygenases from arachidonic acid to less active dihydroxyeicosatrienoic acids. dihydroxyeicosatrienoic acids 173-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 27144772-1 2016 Soluble epoxide hydrolase (sEH) converts highly active epoxyeicosatrienoic acids (EETs) generated by cytochrome P450 (CYP) epoxygenases from arachidonic acid to less active dihydroxyeicosatrienoic acids. dihydroxyeicosatrienoic acids 173-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 27280598-4 2016 OBJECTIVE: To investigate the effects of sodium fusidate on recombinant cytochrome P450 enzymes (1A2, 2C9, 2C19, 2D6 and 3A4) in-vitro. Fusidic Acid 41-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 27245908-1 2016 Cytochrome P450 enzymes are a large family of heme-containing proteins that have important functions in the biotransformation of xenobiotics, including pharmacologic and environmental agents, as well as of endogenously produced chemicals with broad structural and functional diversity. Heme 46-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 27642356-0 2016 Traditional Preparations and Methanol Extracts of Medicinal Plants from Papua New Guinea Exhibit Similar Cytochrome P450 Inhibition. Methanol 29-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 27451096-1 2016 Endothelial and vascular smooth cells generate cytochrome P450 (CYP) arachidonic acid metabolites that can impact endothelial cell function and vascular homeostasis. Arachidonic Acid 69-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 27451096-1 2016 Endothelial and vascular smooth cells generate cytochrome P450 (CYP) arachidonic acid metabolites that can impact endothelial cell function and vascular homeostasis. Arachidonic Acid 69-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. Eicosanoids 38-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. Eicosanoids 38-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. Eicosanoids 38-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. epoxyeicosatrienoic acids 59-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. epoxyeicosatrienoic acids 59-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. epoxyeicosatrienoic acids 59-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. eets 86-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. eets 86-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. eets 86-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. Hydroxyeicosatetraenoic Acids 125-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 27451096-3 2016 The CYP pathway produces two types of eicosanoid products: epoxyeicosatrienoic acids (EETs), formed by CYP epoxygenases, and hydroxyeicosatetraenoic acids (HETEs), formed by CYP hydroxylases. Hydroxyeicosatetraenoic Acids 156-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 27451096-4 2016 Advances in CYP enzymes, EETs, and 20-HETE by pharmacological and genetic means have led to a more complete understanding of how these eicosanoids impact on endothelial cell function. Eicosanoids 135-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 28057170-1 2016 Purpose - The CEIBA cocktail consisting of caffeine (CAF), omeprazole (OZ), dextromethorphan (DM) and losartan (LOS) was previously proposed for the clinical phenotyping of five major human cytochrome P450 (CYP) isoenzymes. ceiba cocktail 14-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-205 28057170-1 2016 Purpose - The CEIBA cocktail consisting of caffeine (CAF), omeprazole (OZ), dextromethorphan (DM) and losartan (LOS) was previously proposed for the clinical phenotyping of five major human cytochrome P450 (CYP) isoenzymes. ceiba cocktail 14-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-210 26831703-0 2016 Facile Synthetic Access to Glycopeptide Antibiotic Precursor Peptides for the Investigation of Cytochrome P450 Action in Glycopeptide Antibiotic Biosynthesis. Glycopeptides 27-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 26247835-2 2016 The polycyclic hydrocarbons (PAHs), pyrene, 1-hydroxypyrene, 1-nitropyrene and 1-acetylpyrene, were found to induce Type I binding spectra with human cytochrome P450 (P450) 2A13 and were converted to various mono- and di-oxygenated products by this enzyme. Hydrocarbons, Cyclic 4-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-177 26747234-0 2016 Cyclooxygenase- and cytochrome P450-derived eicosanoids in stroke. Eicosanoids 44-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 26747234-1 2016 Arachidonic acid (AA) is metabolized by cyclooxygenase (COX) and cytochrome P450 (CYP) enzymes into eicosanoids, which are involved in cardiovascular diseases and stroke. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 26747234-1 2016 Arachidonic acid (AA) is metabolized by cyclooxygenase (COX) and cytochrome P450 (CYP) enzymes into eicosanoids, which are involved in cardiovascular diseases and stroke. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 26747234-1 2016 Arachidonic acid (AA) is metabolized by cyclooxygenase (COX) and cytochrome P450 (CYP) enzymes into eicosanoids, which are involved in cardiovascular diseases and stroke. Eicosanoids 100-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 26747234-1 2016 Arachidonic acid (AA) is metabolized by cyclooxygenase (COX) and cytochrome P450 (CYP) enzymes into eicosanoids, which are involved in cardiovascular diseases and stroke. Eicosanoids 100-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 26153439-11 2016 It was demonstrated that EWD or MXCT pretreatment obviously induced CYP1A2, therefore, in patients taking EWD or MXCT, possible CYP-induced drug interaction should be noted to decrease the risk of therapeutic failure or adverse effects resulting from the use of additional therapeutic agents. mxct 32-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 26153439-11 2016 It was demonstrated that EWD or MXCT pretreatment obviously induced CYP1A2, therefore, in patients taking EWD or MXCT, possible CYP-induced drug interaction should be noted to decrease the risk of therapeutic failure or adverse effects resulting from the use of additional therapeutic agents. mxct 113-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 27239927-3 2016 They initially received dual antiplatelet therapy: clopidogrel 75 mg + acetylsalicylic acid (ASA) 300 mg. Genetic testing was performed in all the patients to reveal the carriage of allelic variants of the genes of cytochrome P-450 isoenzymes and the efficiency of antiplatelet therapy was evaluated. Clopidogrel 51-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-231 27239927-3 2016 They initially received dual antiplatelet therapy: clopidogrel 75 mg + acetylsalicylic acid (ASA) 300 mg. Genetic testing was performed in all the patients to reveal the carriage of allelic variants of the genes of cytochrome P-450 isoenzymes and the efficiency of antiplatelet therapy was evaluated. Aspirin 71-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-231 27239927-3 2016 They initially received dual antiplatelet therapy: clopidogrel 75 mg + acetylsalicylic acid (ASA) 300 mg. Genetic testing was performed in all the patients to reveal the carriage of allelic variants of the genes of cytochrome P-450 isoenzymes and the efficiency of antiplatelet therapy was evaluated. Aspirin 93-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-231 26247835-2 2016 The polycyclic hydrocarbons (PAHs), pyrene, 1-hydroxypyrene, 1-nitropyrene and 1-acetylpyrene, were found to induce Type I binding spectra with human cytochrome P450 (P450) 2A13 and were converted to various mono- and di-oxygenated products by this enzyme. Polycyclic Aromatic Hydrocarbons 29-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-177 26247835-2 2016 The polycyclic hydrocarbons (PAHs), pyrene, 1-hydroxypyrene, 1-nitropyrene and 1-acetylpyrene, were found to induce Type I binding spectra with human cytochrome P450 (P450) 2A13 and were converted to various mono- and di-oxygenated products by this enzyme. pyrene 36-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-177 26247835-2 2016 The polycyclic hydrocarbons (PAHs), pyrene, 1-hydroxypyrene, 1-nitropyrene and 1-acetylpyrene, were found to induce Type I binding spectra with human cytochrome P450 (P450) 2A13 and were converted to various mono- and di-oxygenated products by this enzyme. 1-hydroxypyrene 44-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-177 26247835-2 2016 The polycyclic hydrocarbons (PAHs), pyrene, 1-hydroxypyrene, 1-nitropyrene and 1-acetylpyrene, were found to induce Type I binding spectra with human cytochrome P450 (P450) 2A13 and were converted to various mono- and di-oxygenated products by this enzyme. 1-nitropyrene 61-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-177 26247835-2 2016 The polycyclic hydrocarbons (PAHs), pyrene, 1-hydroxypyrene, 1-nitropyrene and 1-acetylpyrene, were found to induce Type I binding spectra with human cytochrome P450 (P450) 2A13 and were converted to various mono- and di-oxygenated products by this enzyme. 1-Acetylpyrene 79-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-177 26756638-3 2015 Two lesser-known enzymatic pathways of arachidonic acid metabolism are the lipoxygenase (LO) and cytochrome P450 (CYP) pathways. Arachidonic Acid 39-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-112 25659768-6 2015 After 14 days, Amiodarone major oxidative metabolite (mono-N-desethylamiodarone) was detected at limited levels, indicating the presence of active drug metabolism enzymes (i.e. cytochrome P450) in both models. mono-N-desethylamiodarone 54-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-192 26756638-3 2015 Two lesser-known enzymatic pathways of arachidonic acid metabolism are the lipoxygenase (LO) and cytochrome P450 (CYP) pathways. Arachidonic Acid 39-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 26756638-6 2015 Proximal and distal tubular sodium transport and fluid and electrolyte homeostasis are also significantly influenced by renal CYP and LO levels. Sodium 28-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 26593908-2 2015 AAI is detoxified by cytochrome P450 (CYP)-mediated O-demethylation to 8-hydroxyaristolochic acid I (aristolochic acid Ia, AAIa). aristolochic acid I 80-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 26627649-0 2015 Small-angle scattering determination of the shape and localization of human cytochrome P450 embedded in a phospholipid nanodisc environment. Phospholipids 106-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 27747722-10 2015 CONCLUSION: The close relationship between diclofenac and pantoprazole, and the cytochrome P450 and P-glycoprotein systems offers a strong indication that a drug interaction may be occurring. Diclofenac 43-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 26481538-0 2015 Inhibition of human cytochrome P450 isoenzymes by a phenothiazine neuroleptic levomepromazine: An in vitro study. phenothiazine 52-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 26481538-0 2015 Inhibition of human cytochrome P450 isoenzymes by a phenothiazine neuroleptic levomepromazine: An in vitro study. Methotrimeprazine 78-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 26481538-2 2015 The present study was aimed at examining the inhibitory effect of the phenothiazine neuroleptic levomepromazine on main CYP isoenzymes in human liver. phenothiazine 70-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 26481538-2 2015 The present study was aimed at examining the inhibitory effect of the phenothiazine neuroleptic levomepromazine on main CYP isoenzymes in human liver. Methotrimeprazine 96-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 26481538-4 2015 CYP isoenzyme activities were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6) and testosterone 6beta-hydroxylation (CYP3A4). Caffeine 75-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26481538-4 2015 CYP isoenzyme activities were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6) and testosterone 6beta-hydroxylation (CYP3A4). Caffeine 75-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 26481538-4 2015 CYP isoenzyme activities were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6) and testosterone 6beta-hydroxylation (CYP3A4). Diclofenac 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26481538-4 2015 CYP isoenzyme activities were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6) and testosterone 6beta-hydroxylation (CYP3A4). Diclofenac 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 26481538-4 2015 CYP isoenzyme activities were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6) and testosterone 6beta-hydroxylation (CYP3A4). Perazine 150-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26481538-4 2015 CYP isoenzyme activities were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6) and testosterone 6beta-hydroxylation (CYP3A4). bufuralol 186-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26481538-4 2015 CYP isoenzyme activities were determined using the CYP-specific reactions: caffeine 3-N-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), perazine N-demethylation (CYP2C19), bufuralol 1"-hydroxylation (CYP2D6) and testosterone 6beta-hydroxylation (CYP3A4). Testosterone 226-238 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26481538-5 2015 The rates of the CYP-specific reactions were assessed in the absence and presence of levomepromazine (1-50 muM). Methotrimeprazine 85-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 26593908-2 2015 AAI is detoxified by cytochrome P450 (CYP)-mediated O-demethylation to 8-hydroxyaristolochic acid I (aristolochic acid Ia, AAIa). aristolochic acid I 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 26593908-2 2015 AAI is detoxified by cytochrome P450 (CYP)-mediated O-demethylation to 8-hydroxyaristolochic acid I (aristolochic acid Ia, AAIa). aristolochic acid I 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 26593908-2 2015 AAI is detoxified by cytochrome P450 (CYP)-mediated O-demethylation to 8-hydroxyaristolochic acid I (aristolochic acid Ia, AAIa). 8-hydroxyaristolochic acid i 71-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 26593908-2 2015 AAI is detoxified by cytochrome P450 (CYP)-mediated O-demethylation to 8-hydroxyaristolochic acid I (aristolochic acid Ia, AAIa). 8-hydroxyaristolochic acid i 71-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 26396054-2 2015 The objective of the current study was to evaluate the acute and short-term effects of walnut intake on changes in microvascular function and the relationship of these effects to plasma epoxides, the cytochrome-P450-derived metabolites of fatty acids. Epoxy Compounds 186-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-215 27182450-0 2015 CYP-epoxygenase metabolites of docosahexaenoic acid protect HL-1 cardiac cells against LPS-induced cytotoxicity Through SIRT1. Docosahexaenoic Acids 31-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 27182450-5 2015 Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. Docosahexaenoic Acids 82-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 27182450-5 2015 Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. Docosahexaenoic Acids 82-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 27182450-5 2015 Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. epoxydocosapentaenoic acids 87-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 27182450-5 2015 Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. epoxydocosapentaenoic acids 87-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 27182450-5 2015 Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. edps 116-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 27182450-5 2015 Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. edps 116-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 27182450-5 2015 Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. Docosahexaenoic Acids 189-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 27182450-5 2015 Emerging evidence indicates that cytochrome P450 (CYP) epoxygenase metabolites of DHA, epoxydocosapentaenoic acids (EDPs), produce more potent biological activity compared to its precursor DHA. Docosahexaenoic Acids 189-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 27182450-8 2015 Our results illustrate the CYP epoxygenase metabolite of DHA, 19,20-EDP, confers extensive protection to HL-1 cardiac cells against LPS-induced cytotoxicity via activation of SIRT1. Docosahexaenoic Acids 57-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 27182450-8 2015 Our results illustrate the CYP epoxygenase metabolite of DHA, 19,20-EDP, confers extensive protection to HL-1 cardiac cells against LPS-induced cytotoxicity via activation of SIRT1. 19(20)-EpDPE 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 26593908-2 2015 AAI is detoxified by cytochrome P450 (CYP)-mediated O-demethylation to 8-hydroxyaristolochic acid I (aristolochic acid Ia, AAIa). aristolochic acid I 80-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 26593908-2 2015 AAI is detoxified by cytochrome P450 (CYP)-mediated O-demethylation to 8-hydroxyaristolochic acid I (aristolochic acid Ia, AAIa). aaia 123-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 26593908-2 2015 AAI is detoxified by cytochrome P450 (CYP)-mediated O-demethylation to 8-hydroxyaristolochic acid I (aristolochic acid Ia, AAIa). aaia 123-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 26593908-9 2015 We found that the binding orientations of the methoxy group of AAI in binding centers of the CYP enzymes and the energies of AAI binding to the CYP active sites dictate the efficiency of AAI oxidation. aristolochic acid I 63-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 26593908-9 2015 We found that the binding orientations of the methoxy group of AAI in binding centers of the CYP enzymes and the energies of AAI binding to the CYP active sites dictate the efficiency of AAI oxidation. aristolochic acid I 63-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-147 26593908-9 2015 We found that the binding orientations of the methoxy group of AAI in binding centers of the CYP enzymes and the energies of AAI binding to the CYP active sites dictate the efficiency of AAI oxidation. aristolochic acid I 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 26593908-9 2015 We found that the binding orientations of the methoxy group of AAI in binding centers of the CYP enzymes and the energies of AAI binding to the CYP active sites dictate the efficiency of AAI oxidation. aristolochic acid I 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-147 26593908-9 2015 We found that the binding orientations of the methoxy group of AAI in binding centers of the CYP enzymes and the energies of AAI binding to the CYP active sites dictate the efficiency of AAI oxidation. aristolochic acid I 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 26593908-9 2015 We found that the binding orientations of the methoxy group of AAI in binding centers of the CYP enzymes and the energies of AAI binding to the CYP active sites dictate the efficiency of AAI oxidation. aristolochic acid I 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-147 26593908-10 2015 Our results indicate that utilization of experimental and theoretical methods is an appropriate study design to examine the CYP-catalyzed reaction mechanisms of AAI oxidation and contributions of human hepatic CYPs to this metabolism. aristolochic acid I 161-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 26276083-0 2015 Toxicokinetics of lefetamine and derived diphenylethylamine designer drugs-Contribution of human cytochrome P450 isozymes to their main phase I metabolic steps. lephetamine 18-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-112 26443405-9 2015 CYP2D6 and CYP3A4 played an important role in the isomerization and glycination of limonoids in HLMs, whereas other CYP isoforms were considerably less active. Limonins 83-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26452461-7 2015 Cytochrome P450 activity was delivered into Human cervix carcinoma cells via transfecting P22-CYP nanoparticles with lipofectamine. Lipofectamine 117-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26318115-2 2015 Paraoxonase-1 (PON1), butyrylcholinesterase (BChE) and Cytochrome-P450 constitute major classes of XME involved in the detoxification of pesticide chemicals, in particular organophosphates. XME 99-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 26318115-2 2015 Paraoxonase-1 (PON1), butyrylcholinesterase (BChE) and Cytochrome-P450 constitute major classes of XME involved in the detoxification of pesticide chemicals, in particular organophosphates. Organophosphates 172-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 25940755-3 2015 CYP activity was assessed by the apparent clearance of midazolam (CYP3A), omeprazole/5-hydroxyomeprazol ratio in plasma (CYP2C19) and losartan/E3174 ratio in urine (CYP2C9). Midazolam 55-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 25940755-3 2015 CYP activity was assessed by the apparent clearance of midazolam (CYP3A), omeprazole/5-hydroxyomeprazol ratio in plasma (CYP2C19) and losartan/E3174 ratio in urine (CYP2C9). Omeprazole 74-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 25940755-3 2015 CYP activity was assessed by the apparent clearance of midazolam (CYP3A), omeprazole/5-hydroxyomeprazol ratio in plasma (CYP2C19) and losartan/E3174 ratio in urine (CYP2C9). 5-hydroxyomeprazol 85-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 25940755-3 2015 CYP activity was assessed by the apparent clearance of midazolam (CYP3A), omeprazole/5-hydroxyomeprazol ratio in plasma (CYP2C19) and losartan/E3174 ratio in urine (CYP2C9). Losartan 134-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 25940755-3 2015 CYP activity was assessed by the apparent clearance of midazolam (CYP3A), omeprazole/5-hydroxyomeprazol ratio in plasma (CYP2C19) and losartan/E3174 ratio in urine (CYP2C9). losartan carboxylic acid 143-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26220948-0 2015 Human Liver Cytochrome P450 Enzymes and Microsomal Thiol Methyltransferase Are Involved in the Stereoselective Formation and Methylation of the Pharmacologically Active Metabolite of Clopidogrel. Clopidogrel 183-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 26350273-2 2015 Hydrocodone undergoes cytochrome P-450 (CYP)-mediated metabolism involving the CYP3A4 and CYP2D6 isozymes. Hydrocodone 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 26350273-2 2015 Hydrocodone undergoes cytochrome P-450 (CYP)-mediated metabolism involving the CYP3A4 and CYP2D6 isozymes. Hydrocodone 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 26214067-0 2015 Polymorphisms in CYP-mediated arachidonic acid routes affect the outcome of renal transplantation. Arachidonic Acid 30-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Toremifene 102-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Toremifene 102-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Toremifene 102-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 271-274 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Toremifene 102-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 271-274 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. n-desmethyl 109-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. n-desmethyl 109-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. ndm 122-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. ndm 122-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Estetrol 139-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Estetrol 139-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Estetrol 148-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Estetrol 148-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 26423799-3 2015 In this study, we examined the role of individual cytochrome P450 (CYP) isoforms in the metabolism of TOR to N-desmethyl (NDM), 4-hydroxy (4OH) and 4OH-NDM metabolites in comparison with TAM using human liver microsomes (HLMs) with selective chemical inhibitors for each CYP isoform and recombinant CYP proteins. Tamoxifen 187-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 26214067-1 2015 BACKGROUND: Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP) enzymes to vasoactive metabolites (mainly epoxyeicosatrienoic acids) which are known to play a protective role against damaging processes that may occur after re-oxygenation of the graft. Arachidonic Acid 12-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 26214067-1 2015 BACKGROUND: Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP) enzymes to vasoactive metabolites (mainly epoxyeicosatrienoic acids) which are known to play a protective role against damaging processes that may occur after re-oxygenation of the graft. Arachidonic Acid 12-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 26214067-1 2015 BACKGROUND: Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP) enzymes to vasoactive metabolites (mainly epoxyeicosatrienoic acids) which are known to play a protective role against damaging processes that may occur after re-oxygenation of the graft. epoxyeicosatrienoic acids 116-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 26214067-1 2015 BACKGROUND: Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP) enzymes to vasoactive metabolites (mainly epoxyeicosatrienoic acids) which are known to play a protective role against damaging processes that may occur after re-oxygenation of the graft. epoxyeicosatrienoic acids 116-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 26100226-0 2015 Inhibition of human cytochrome P450 enzymes by licochalcone A, a naturally occurring constituent of licorice. licochalcone A 47-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 26106235-0 2015 Novel Bioactivation Pathway of Benzbromarone Mediated by Cytochrome P450. Benzbromarone 31-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 26135009-3 2015 In this study we assessed the perpetrator potential of desloratadine based on in vitro studies of its inhibitory effects on cytochrome P450 and UDP-glucuronosyltransferase (UGT) enzymes in human liver microsomes (HLM). desloratadine 55-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 26336590-0 2015 Measurement of Human Cytochrome P450 Enzyme Induction Based on Mesalazine and Mosapride Citrate Treatments Using a Luminescent Assay. Mesalamine 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 26336590-0 2015 Measurement of Human Cytochrome P450 Enzyme Induction Based on Mesalazine and Mosapride Citrate Treatments Using a Luminescent Assay. mosapride 78-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 25920956-5 2015 Patients were genotyped for polymorphic variants and cytochrome P450 (CYP)-eicosanoid CSF levels were measured over 14 days. Eicosanoids 75-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 26163112-0 2015 Role of cytochrome P450 and UDP-glucuronosyltransferases in metabolic pathway of homoegonol in human liver microsomes. homoegonol 81-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). Irinotecan 272-282 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). Irinotecan 272-282 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). Fluorouracil 284-298 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). Fluorouracil 284-298 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). Leucovorin 304-314 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). Leucovorin 304-314 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). FOLFIRI regimen 316-323 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 25934339-1 2015 The purpose of this study is to evaluate the influence of germline polymorphisms of cytochrome P450 (CYP450) on objective response, progression-free survival (PFS) and overall suruvival (OS) in metastatic colorectal cancer (mCRC) receiving the combination chemotherapy of irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI). FOLFIRI regimen 316-323 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 25934339-2 2015 All SNPs in CYP450, whose minor allele frequency were more than 10 %, were genotyped in 82 patients with mCRC who received first-line FOLFIRI regimen. FOLFIRI regimen 134-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-18 26157146-8 2015 Previous studies showed that human cytochrome P450 (P450) 2D6 can catalyze thebaine O(3)-demethylation. Thebaine 75-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-61 25940823-3 2015 Tamoxifen is bioactivated by cytochrome P450 (CYP) enzymes such as CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP3A4/5, resulting in the formation of active metabolites, including 4-hydroxy-tamoxifen and endoxifen. Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 25940823-3 2015 Tamoxifen is bioactivated by cytochrome P450 (CYP) enzymes such as CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP3A4/5, resulting in the formation of active metabolites, including 4-hydroxy-tamoxifen and endoxifen. Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 26489070-5 2015 On the other hand, dosage adjustments may be required when vortioxetine is coadministered with strong CYP2D6 inhibitors or broad-spectrum CYP inducers. Vortioxetine 59-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 25940823-3 2015 Tamoxifen is bioactivated by cytochrome P450 (CYP) enzymes such as CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP3A4/5, resulting in the formation of active metabolites, including 4-hydroxy-tamoxifen and endoxifen. 4'-hydroxytamoxifen 173-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 25940823-3 2015 Tamoxifen is bioactivated by cytochrome P450 (CYP) enzymes such as CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP3A4/5, resulting in the formation of active metabolites, including 4-hydroxy-tamoxifen and endoxifen. 4'-hydroxytamoxifen 173-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 25940823-3 2015 Tamoxifen is bioactivated by cytochrome P450 (CYP) enzymes such as CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP3A4/5, resulting in the formation of active metabolites, including 4-hydroxy-tamoxifen and endoxifen. 4-hydroxy-N-desmethyltamoxifen 197-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 25940823-3 2015 Tamoxifen is bioactivated by cytochrome P450 (CYP) enzymes such as CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP3A4/5, resulting in the formation of active metabolites, including 4-hydroxy-tamoxifen and endoxifen. 4-hydroxy-N-desmethyltamoxifen 197-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 26015560-7 2015 In an in vitro cytochrome P450 (CYP) panel, cabozantinib and EXEL-1644 both inhibited most potently CYP2C8 (Kiapp = 4.6 and 1.1 microM, respectively). cabozantinib 44-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-30 26015560-7 2015 In an in vitro cytochrome P450 (CYP) panel, cabozantinib and EXEL-1644 both inhibited most potently CYP2C8 (Kiapp = 4.6 and 1.1 microM, respectively). cabozantinib 44-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 26015560-7 2015 In an in vitro cytochrome P450 (CYP) panel, cabozantinib and EXEL-1644 both inhibited most potently CYP2C8 (Kiapp = 4.6 and 1.1 microM, respectively). UNII-NI7FZ38DO2 61-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-30 26015560-7 2015 In an in vitro cytochrome P450 (CYP) panel, cabozantinib and EXEL-1644 both inhibited most potently CYP2C8 (Kiapp = 4.6 and 1.1 microM, respectively). UNII-NI7FZ38DO2 61-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 26163112-4 2015 We characterized the cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzymes responsible for homoegonol metabolism using human liver microsomes, and cDNA-expressed CYP and UGT enzymes. homoegonol 105-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 26163112-4 2015 We characterized the cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzymes responsible for homoegonol metabolism using human liver microsomes, and cDNA-expressed CYP and UGT enzymes. homoegonol 105-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 26622646-10 2015 Additional large-scale studies are required in order to evaluate the role of CYP enzymes in bortezomib treatments for patients with multiple myeloma. Bortezomib 92-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 26104032-2 2015 Pharmacokinetic studies show that pitavastatin is carried into the liver by a range of transporters and is minimally metabolized by cytochrome P450 in healthy volunteers, indicating a reduced potential for drug-drug interactions (DDIs). pitavastatin 34-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-147 25808545-6 2015 Differentiated cells produce albumin and apolipoprotein B100 at levels equivalent to primary human hepatocytes, while demonstrating an 8-fold induction of CYP450 activity in response to aryl hydrocarbon receptor (AhR) agonist omeprazole and a 10-fold induction in response to PXR agonist rifampicin. Omeprazole 226-236 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-161 26181717-0 2015 The Effect of 5-Aminolevulinic Acid on Cytochrome P450-Mediated Prodrug Activation. 5-amino levulinic acid 14-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 26181717-4 2015 We hypothesized that the addition of 5-aminolevulinic acid (ALA), a precursor in the porphyrin biosynthetic pathway, enhances the synthesis of heme, leading to the up-regulation of CYP activity. 5-amino levulinic acid 37-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-184 26181717-4 2015 We hypothesized that the addition of 5-aminolevulinic acid (ALA), a precursor in the porphyrin biosynthetic pathway, enhances the synthesis of heme, leading to the up-regulation of CYP activity. 5-amino levulinic acid 60-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-184 26181717-4 2015 We hypothesized that the addition of 5-aminolevulinic acid (ALA), a precursor in the porphyrin biosynthetic pathway, enhances the synthesis of heme, leading to the up-regulation of CYP activity. Porphyrins 85-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-184 26181717-4 2015 We hypothesized that the addition of 5-aminolevulinic acid (ALA), a precursor in the porphyrin biosynthetic pathway, enhances the synthesis of heme, leading to the up-regulation of CYP activity. Heme 143-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-184 26181717-7 2015 We show here that ALA increased CYP2A6-dependent tegafur activation, suggesting that ALA elevated CYP activity and potentiated the activation of the prodrug. 5-amino levulinic acid 18-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 26181717-7 2015 We show here that ALA increased CYP2A6-dependent tegafur activation, suggesting that ALA elevated CYP activity and potentiated the activation of the prodrug. 5-amino levulinic acid 85-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 25808545-6 2015 Differentiated cells produce albumin and apolipoprotein B100 at levels equivalent to primary human hepatocytes, while demonstrating an 8-fold induction of CYP450 activity in response to aryl hydrocarbon receptor (AhR) agonist omeprazole and a 10-fold induction in response to PXR agonist rifampicin. Rifampin 288-298 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-161 26056268-3 2015 Here we identified the cytochrome P450 gene CYP4F22 (cytochrome P450, family 4, subfamily F, polypeptide 22) as the long-sought fatty acid omega-hydroxylase gene required for acylceramide production. acylceramide 175-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-78 25986600-2 2015 Their antihypertensive action may be due to the reduction of the omega-6/omega-3 ratio and the resulting competitive effect of omega-3 as compared to arachidonic acid (an omega-6 PUFA) as a substrate of cytochrome P450 (CYP450) enzymes involved in the production of vasoactive mediators. omega-3 127-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 203-218 25986600-2 2015 Their antihypertensive action may be due to the reduction of the omega-6/omega-3 ratio and the resulting competitive effect of omega-3 as compared to arachidonic acid (an omega-6 PUFA) as a substrate of cytochrome P450 (CYP450) enzymes involved in the production of vasoactive mediators. omega-3 127-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 220-226 25834924-5 2015 Mechanisms through which omega-3 FA act are manifolds and still a matter of investigation: beside their interaction with ion channel and their influence on plasma membrane fluidity, probably the main effect is acting as competitor for cytochrome P-450 (CYP) with respect to omega-6 FA. omega-6 fa 274-284 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 235-251 25834924-5 2015 Mechanisms through which omega-3 FA act are manifolds and still a matter of investigation: beside their interaction with ion channel and their influence on plasma membrane fluidity, probably the main effect is acting as competitor for cytochrome P-450 (CYP) with respect to omega-6 FA. omega-6 fa 274-284 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 253-256 25862215-0 2015 Lisdexamfetamine Dimesylate Effects on the Pharmacokinetics of Cytochrome P450 Substrates in Healthy Adults in an Open-Label, Randomized, Crossover Study. Lisdexamfetamine Dimesylate 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 26042637-1 2015 The remarkable aliphatic C-H hydroxylations catalyzed by the heme-containing enzyme, cytochrome P450, have attracted sustained attention for more than four decades. Heme 61-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 26047326-0 2015 All-Trans Retinoic Acid Activity in Acute Myeloid Leukemia: Role of Cytochrome P450 Enzyme Expression by the Microenvironment. Tretinoin 10-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 26082767-3 2015 We have recently shown the possible role of cytochrome P450 (CYP) in smoking/nicotine-mediated viral replication. Nicotine 77-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 26082767-3 2015 We have recently shown the possible role of cytochrome P450 (CYP) in smoking/nicotine-mediated viral replication. Nicotine 77-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 26082767-4 2015 In this review, we focus on the potential role of CYP pathway in polycyclic aromatic hydrocarbons (PAH), important constituents of cigarette smoke, mediated HIV pathogenesis. Polycyclic Aromatic Hydrocarbons 65-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 26082767-4 2015 In this review, we focus on the potential role of CYP pathway in polycyclic aromatic hydrocarbons (PAH), important constituents of cigarette smoke, mediated HIV pathogenesis. Polycyclic Aromatic Hydrocarbons 99-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 26082767-5 2015 More specifically, we will discuss the role of CYP1A1 and CYP1B1, which are the major PAH-activating CYP enzymes. Polycyclic Aromatic Hydrocarbons 86-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 25862215-1 2015 INTRODUCTION: This open-label, randomized, two-period drug interaction study assessed lisdexamfetamine dimesylate (LDX) effects on cytochrome P450 (CYP) enzyme (CYP1A2, CYP2D6, CYP2C19, and CYP3A) activity. Lisdexamfetamine Dimesylate 86-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 25862215-1 2015 INTRODUCTION: This open-label, randomized, two-period drug interaction study assessed lisdexamfetamine dimesylate (LDX) effects on cytochrome P450 (CYP) enzyme (CYP1A2, CYP2D6, CYP2C19, and CYP3A) activity. Lisdexamfetamine Dimesylate 86-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 25808509-0 2015 Association of CYP450 single nucleotide polymorphisms with the efficacy of epidural ropivacaine during mastectomy. Ropivacaine 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-21 25850753-1 2015 Tacrolimus is prescribed to prevent allograft rejection in pediatric liver transplant recipients; however, its metabolism through the cytochrome P-450 enzyme system presents a multitude of challenges in regard to drug interactions. Tacrolimus 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 25897563-7 2015 According to the results obtained in the heterogeneous process, Cu4CuP3S mimicked the activity of cytochrome P-450 and catecholase. cu4cup3s 64-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-130 25603423-3 2015 In addition, many tumors over-express cyclooxygenase, lipoxygenase or cytochrome P450 enzymes that mediate the biotransformation of omega-6 polyunsaturated fatty acids (PUFAs) to potent eicosanoid regulators of tumor cell proliferation and cell death. omega-6 polyunsaturated fatty acids 132-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 25603423-3 2015 In addition, many tumors over-express cyclooxygenase, lipoxygenase or cytochrome P450 enzymes that mediate the biotransformation of omega-6 polyunsaturated fatty acids (PUFAs) to potent eicosanoid regulators of tumor cell proliferation and cell death. Fatty Acids, Unsaturated 169-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 25603423-3 2015 In addition, many tumors over-express cyclooxygenase, lipoxygenase or cytochrome P450 enzymes that mediate the biotransformation of omega-6 polyunsaturated fatty acids (PUFAs) to potent eicosanoid regulators of tumor cell proliferation and cell death. Eicosanoids 186-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 25808509-1 2015 BACKGROUND: Ropivacaine is frequently used for local anesthesia in the clinic and is metabolized by cytochrome P450 (CYP450) in the liver. Ropivacaine 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 25808509-1 2015 BACKGROUND: Ropivacaine is frequently used for local anesthesia in the clinic and is metabolized by cytochrome P450 (CYP450) in the liver. Ropivacaine 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-123 25808509-3 2015 In this study, we selected six CYP450 polymorphisms from the dbSNP and HapMap databases, using a combination of functional analysis and Tag SNP strategies and examined these polymorphisms for association with the efficacy of epidural ropivacaine in patients during mastectomy. Ropivacaine 234-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-37 25677220-1 2015 Cyclophosphamide (Cy) is a prodrug that depends on bioactivation by hepatic cytochrome P450 (CYP) enzymes for its cytotoxicity. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 25900452-1 2015 Thromboxane synthase (CYP5A1) is a non-classical cytochrome P450 (CYP) expressed in human platelets that mediates vascular homeostasis by producing thromboxane A2 (TXA2) through the isomerization of prostaglandin H2 (PGH2). Thromboxane A2 148-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 25900452-1 2015 Thromboxane synthase (CYP5A1) is a non-classical cytochrome P450 (CYP) expressed in human platelets that mediates vascular homeostasis by producing thromboxane A2 (TXA2) through the isomerization of prostaglandin H2 (PGH2). Thromboxane A2 148-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 25900452-1 2015 Thromboxane synthase (CYP5A1) is a non-classical cytochrome P450 (CYP) expressed in human platelets that mediates vascular homeostasis by producing thromboxane A2 (TXA2) through the isomerization of prostaglandin H2 (PGH2). Thromboxane A2 164-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 25900452-1 2015 Thromboxane synthase (CYP5A1) is a non-classical cytochrome P450 (CYP) expressed in human platelets that mediates vascular homeostasis by producing thromboxane A2 (TXA2) through the isomerization of prostaglandin H2 (PGH2). Thromboxane A2 164-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 25900452-1 2015 Thromboxane synthase (CYP5A1) is a non-classical cytochrome P450 (CYP) expressed in human platelets that mediates vascular homeostasis by producing thromboxane A2 (TXA2) through the isomerization of prostaglandin H2 (PGH2). Prostaglandin H2 199-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 25900452-1 2015 Thromboxane synthase (CYP5A1) is a non-classical cytochrome P450 (CYP) expressed in human platelets that mediates vascular homeostasis by producing thromboxane A2 (TXA2) through the isomerization of prostaglandin H2 (PGH2). Prostaglandin H2 199-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 25900452-1 2015 Thromboxane synthase (CYP5A1) is a non-classical cytochrome P450 (CYP) expressed in human platelets that mediates vascular homeostasis by producing thromboxane A2 (TXA2) through the isomerization of prostaglandin H2 (PGH2). Prostaglandin H2 217-221 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 25900452-1 2015 Thromboxane synthase (CYP5A1) is a non-classical cytochrome P450 (CYP) expressed in human platelets that mediates vascular homeostasis by producing thromboxane A2 (TXA2) through the isomerization of prostaglandin H2 (PGH2). Prostaglandin H2 217-221 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 25677220-1 2015 Cyclophosphamide (Cy) is a prodrug that depends on bioactivation by hepatic cytochrome P450 (CYP) enzymes for its cytotoxicity. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 25677220-1 2015 Cyclophosphamide (Cy) is a prodrug that depends on bioactivation by hepatic cytochrome P450 (CYP) enzymes for its cytotoxicity. Cyclophosphamide 0-2 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 25677220-1 2015 Cyclophosphamide (Cy) is a prodrug that depends on bioactivation by hepatic cytochrome P450 (CYP) enzymes for its cytotoxicity. Cyclophosphamide 0-2 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 25585969-6 2015 RESULTS: Pharmacogenetic testing revealed a relation between this adverse event and an allelic variant of cytochrome P450, CYP2C9, subsequently leading to discontinuation of the drug along with counseling to caution the patient to avoid the use of celecoxib and other drugs metabolized by the same enzyme. Celecoxib 248-257 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 25515945-1 2015 Voriconazole is frequently utilized for the prevention and treatment of invasive fungal infections (IFIs), and is extensively metabolized by the cytochrome P450 (CYP) system. Voriconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 25470432-0 2015 Metronidazole reduces the expression of cytochrome P450 enzymes in HepaRG cells and cryopreserved human hepatocytes. Metronidazole 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 25470432-12 2015 We also found that MTZ use may alter the disposition of drugs metabolized by the CYP isozymes investigated. Metronidazole 19-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 25793416-1 2015 Cytochrome P450 enzymes are renowned for their ability to insert oxygen into an enormous variety of compounds with a high degree of chemo- and regio-selectivity under mild conditions. Oxygen 65-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 25515945-1 2015 Voriconazole is frequently utilized for the prevention and treatment of invasive fungal infections (IFIs), and is extensively metabolized by the cytochrome P450 (CYP) system. Voriconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 25162212-1 2015 BACKGROUND: Tacrolimus (TAC) is a known substrate for cytochrome P450 (CYP) enzyme. Tacrolimus 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 25162212-1 2015 BACKGROUND: Tacrolimus (TAC) is a known substrate for cytochrome P450 (CYP) enzyme. Tacrolimus 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 25658971-2 2015 This study presents density functional theory calculations of the CYP-mediated metabolism of acetaminophen and a series of related compounds that can form reactive metabolites by hydrogen abstraction. Acetaminophen 93-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 25658971-2 2015 This study presents density functional theory calculations of the CYP-mediated metabolism of acetaminophen and a series of related compounds that can form reactive metabolites by hydrogen abstraction. Hydrogen 179-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 29560240-1 2015 Cytochrome P450 enzymes are heme based monoxygenases that catalyse a range of oxygen atom transfer reactions with various substrates, including aliphatic and aromatic hydroxylation as well as epoxidation reactions. Oxygen 42-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 25642975-0 2015 Oxidation of Acenaphthene and Acenaphthylene by Human Cytochrome P450 Enzymes. acenaphthene 13-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 25642975-0 2015 Oxidation of Acenaphthene and Acenaphthylene by Human Cytochrome P450 Enzymes. acenaphthylene 30-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 25253429-0 2015 MDR1 and cytochrome P450 gene-expression profiles as markers of chemosensitivity in human chronic myelogenous leukemia cells treated with cisplatin and Ru(III) metallocomplexes. Cisplatin 138-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-24 25253429-0 2015 MDR1 and cytochrome P450 gene-expression profiles as markers of chemosensitivity in human chronic myelogenous leukemia cells treated with cisplatin and Ru(III) metallocomplexes. ru(iii) 152-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-24 25388680-2 2015 We investigated the relationship between polymorphic genes involved in EET biosynthesis/metabolism, cytochrome P450 (CYP) eicosanoid levels, and outcomes in 363 patients with aneurysmal subarachnoid hemorrhage (aSAH). Eicosanoids 122-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-120 26002730-2 2015 CYP enzymes catalyze monooxygenation reactions by inserting one oxygen atom from O2 into an enormous number and variety of substrates. Oxygen 25-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 25533794-7 2015 Among CYP isoforms, CYP2E1 activity was selectively inhibited by MDGA through a competitive inhibitory mode, with an inhibitory constant (Ki) value of 13.1 microM. dihydroguaiaretic acid 65-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 25636872-0 2015 Investigation of cytochrome P450 inhibitory properties of maslinic acid, a bioactive compound from Olea europaea L., and its structure-activity relationship. maslinic acid 58-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-32 25448283-0 2015 Activation of the anticancer drugs cyclophosphamide and ifosfamide by cytochrome P450 BM3 mutants. Cyclophosphamide 35-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 25448283-0 2015 Activation of the anticancer drugs cyclophosphamide and ifosfamide by cytochrome P450 BM3 mutants. Ifosfamide 56-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 25448283-1 2015 Cyclophosphamide (CPA) and ifosfamide (IFA) are widely used anticancer agents that require metabolic activation by cytochrome P450 (CYP) enzymes. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-130 25448283-1 2015 Cyclophosphamide (CPA) and ifosfamide (IFA) are widely used anticancer agents that require metabolic activation by cytochrome P450 (CYP) enzymes. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 25448283-1 2015 Cyclophosphamide (CPA) and ifosfamide (IFA) are widely used anticancer agents that require metabolic activation by cytochrome P450 (CYP) enzymes. Cyclophosphamide 18-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-130 25448283-1 2015 Cyclophosphamide (CPA) and ifosfamide (IFA) are widely used anticancer agents that require metabolic activation by cytochrome P450 (CYP) enzymes. Cyclophosphamide 18-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 25448283-1 2015 Cyclophosphamide (CPA) and ifosfamide (IFA) are widely used anticancer agents that require metabolic activation by cytochrome P450 (CYP) enzymes. Ifosfamide 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-130 25448283-1 2015 Cyclophosphamide (CPA) and ifosfamide (IFA) are widely used anticancer agents that require metabolic activation by cytochrome P450 (CYP) enzymes. Ifosfamide 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 25448283-1 2015 Cyclophosphamide (CPA) and ifosfamide (IFA) are widely used anticancer agents that require metabolic activation by cytochrome P450 (CYP) enzymes. Ifosfamide 39-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-130 25448283-1 2015 Cyclophosphamide (CPA) and ifosfamide (IFA) are widely used anticancer agents that require metabolic activation by cytochrome P450 (CYP) enzymes. Ifosfamide 39-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 25448283-3 2015 Gene-directed enzyme prodrug therapies (GDEPT) have been suggested to facilitate local CPA and IFA bioactivation by expressing CYP enzymes within the tumor cells, thereby increasing efficacy. Cyclophosphamide 87-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 25448283-4 2015 We screened bacterial CYP BM3 mutants, previously engineered to metabolize drug-like compounds, for their ability to catalyze 4-hydroxylation of CPA and IFA. Cyclophosphamide 145-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 25448283-4 2015 We screened bacterial CYP BM3 mutants, previously engineered to metabolize drug-like compounds, for their ability to catalyze 4-hydroxylation of CPA and IFA. Ifosfamide 153-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 25448283-5 2015 Two CYP BM3 mutants showed very rapid initial bioactivation of CPA and IFA, followed by a slower phase of product formation. Cyclophosphamide 63-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 25448283-7 2015 Using purified CYP BM3 as an extracellular bioactivation tool, cytotoxicity of CPA and IFA metabolism was confirmed in U2OS cells. Cyclophosphamide 79-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 25448283-9 2015 To our knowledge, the observed rate of CPA and IFA 4-hydroxylation by these CYP BM3 mutants is the fastest reported to date, and might be of potential interest for CPA and IFA GDEPT. Cyclophosphamide 39-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 25448283-9 2015 To our knowledge, the observed rate of CPA and IFA 4-hydroxylation by these CYP BM3 mutants is the fastest reported to date, and might be of potential interest for CPA and IFA GDEPT. Cyclophosphamide 164-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 26002730-2 2015 CYP enzymes catalyze monooxygenation reactions by inserting one oxygen atom from O2 into an enormous number and variety of substrates. Oxygen 81-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-3 2015 The catalytic versatility of CYP stems from its ability to functionalize unactivated carbon-hydrogen (C-H) bonds of substrates through monooxygenation. hydrogen carbon 85-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 26002730-5 2015 CYP-mediated hydroxylations occur via a consensus H atom abstraction/oxygen rebound mechanism involving an initial abstraction by CpdI of a H atom from the substrate, generating a highly-reactive protonated Compound II (CpdII) intermediate (FeIV-OH) and a carbon-centered alkyl radical that rebounds onto the ferryl hydroxyl moiety to yield the hydroxylated substrate. Oxygen 69-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-5 2015 CYP-mediated hydroxylations occur via a consensus H atom abstraction/oxygen rebound mechanism involving an initial abstraction by CpdI of a H atom from the substrate, generating a highly-reactive protonated Compound II (CpdII) intermediate (FeIV-OH) and a carbon-centered alkyl radical that rebounds onto the ferryl hydroxyl moiety to yield the hydroxylated substrate. feiv-oh 241-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26233909-4 2015 CYP4F3A in myeloid tissue catalyzes the omega-hydroxylation of leukotriene B4 to 20-hydroxy leukotriene B4, an inactivation process that is critical for the regulation of the inflammatory response. Leukotrienes 63-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 26233909-4 2015 CYP4F3A in myeloid tissue catalyzes the omega-hydroxylation of leukotriene B4 to 20-hydroxy leukotriene B4, an inactivation process that is critical for the regulation of the inflammatory response. 5,12,20-trihydroxy-6,8,10,14-eicosatetraenoic acid 81-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 26233910-2 2015 Some cytochrome P450 (CYP) enzymes play key roles in bile acid synthesis. Bile Acids and Salts 53-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-20 26233903-0 2015 Involvement of Cytochrome P450 in Reactive Oxygen Species Formation and Cancer. Reactive Oxygen Species 34-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-30 26233903-1 2015 This review examines the involvement of cytochrome P450 (CYP) enzymes in the formation of reactive oxygen species in biological systems and discusses the possible involvement of reactive oxygen species and CYP enzymes in cancer. Reactive Oxygen Species 90-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 26233903-1 2015 This review examines the involvement of cytochrome P450 (CYP) enzymes in the formation of reactive oxygen species in biological systems and discusses the possible involvement of reactive oxygen species and CYP enzymes in cancer. Reactive Oxygen Species 90-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 26233910-2 2015 Some cytochrome P450 (CYP) enzymes play key roles in bile acid synthesis. Bile Acids and Salts 53-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 26002730-5 2015 CYP-mediated hydroxylations occur via a consensus H atom abstraction/oxygen rebound mechanism involving an initial abstraction by CpdI of a H atom from the substrate, generating a highly-reactive protonated Compound II (CpdII) intermediate (FeIV-OH) and a carbon-centered alkyl radical that rebounds onto the ferryl hydroxyl moiety to yield the hydroxylated substrate. Carbon 256-262 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26233903-1 2015 This review examines the involvement of cytochrome P450 (CYP) enzymes in the formation of reactive oxygen species in biological systems and discusses the possible involvement of reactive oxygen species and CYP enzymes in cancer. Reactive Oxygen Species 178-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 26233903-3 2015 Two endogenous sources of reactive oxygen species are the mammalian CYP-dependent microsomal electron transport system and the mitochondrial electron transport chain. Reactive Oxygen Species 26-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 26233903-4 2015 CYP enzymes catalyze the oxygenation of an organic substrate and the simultaneous reduction of molecular oxygen. Oxygen 25-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26233905-3 2015 In polychemotherapy, prodrugs (like ifosfamide), which have to be activated by cytochrome P450 enzymes (CYPs), play an important role. Ifosfamide 36-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 26002730-5 2015 CYP-mediated hydroxylations occur via a consensus H atom abstraction/oxygen rebound mechanism involving an initial abstraction by CpdI of a H atom from the substrate, generating a highly-reactive protonated Compound II (CpdII) intermediate (FeIV-OH) and a carbon-centered alkyl radical that rebounds onto the ferryl hydroxyl moiety to yield the hydroxylated substrate. centered alkyl radical 263-285 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. Hydrogen Peroxide 20-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. peracids 36-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. perborate 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. sodium percarbonate 57-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. metaperiodate 71-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. chlorite 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. iodosobenzene 92-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26394652-0 2015 Cytochrome P450 Isoforms in the Metabolism of Decursin and Decursinol Angelate from Korean Angelica. decursin 59-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. n-oxides 110-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. Oxygen 132-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). Pyranocoumarins 65-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 26002730-6 2015 CYP enzymes utilize hydroperoxides, peracids, perborate, percarbonate, periodate, chlorite, iodosobenzene and N-oxides as surrogate oxygen atom donors to oxygenate substrates via the shunt pathway in the absence of NAD(P)H/O2 and reduction-oxidation (redox) auxiliary proteins. Oxygen 223-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). Pyranocoumarins 65-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). Pyranocoumarins 65-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-265 26002734-1 2015 Hepatic microsomal cytochrome P450 (CYP) enzymes have broad and overlapping substrate specificity and catalyze a variety of monooxygenase reactions, including aliphatic and aromatic hydroxylations, N-hydroxylations, oxygenations of heteroatoms (N, S, P and I), alkene and arene epoxidations, dehalogenations, dehydrogenations and N-, O- and S-dealkylations. Alkenes 261-267 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). decursin 89-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 26002734-1 2015 Hepatic microsomal cytochrome P450 (CYP) enzymes have broad and overlapping substrate specificity and catalyze a variety of monooxygenase reactions, including aliphatic and aromatic hydroxylations, N-hydroxylations, oxygenations of heteroatoms (N, S, P and I), alkene and arene epoxidations, dehalogenations, dehydrogenations and N-, O- and S-dealkylations. Alkenes 261-267 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). decursin 89-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). decursin 89-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-265 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). decursin 110-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). decursin 110-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). decursin 110-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-265 26002734-1 2015 Hepatic microsomal cytochrome P450 (CYP) enzymes have broad and overlapping substrate specificity and catalyze a variety of monooxygenase reactions, including aliphatic and aromatic hydroxylations, N-hydroxylations, oxygenations of heteroatoms (N, S, P and I), alkene and arene epoxidations, dehalogenations, dehydrogenations and N-, O- and S-dealkylations. arene 272-277 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). decursin 89-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). decursin 89-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). decursin 89-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-265 26002734-1 2015 Hepatic microsomal cytochrome P450 (CYP) enzymes have broad and overlapping substrate specificity and catalyze a variety of monooxygenase reactions, including aliphatic and aromatic hydroxylations, N-hydroxylations, oxygenations of heteroatoms (N, S, P and I), alkene and arene epoxidations, dehalogenations, dehydrogenations and N-, O- and S-dealkylations. arene 272-277 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 26002735-0 2015 Cytochrome p450 enzymes in the bioactivation of polyunsaturated Fatty acids and their role in cardiovascular disease. Fatty Acids, Unsaturated 48-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). doh 129-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 26002735-1 2015 Various members of the cytochrome P450 (CYP) superfamily have the capacity of metabolizing omega-6 and omega-3 polyunsaturated fatty acids (n-6 and n-3 PUFAs). omega-6 91-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). doh 129-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). doh 129-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-265 26002735-1 2015 Various members of the cytochrome P450 (CYP) superfamily have the capacity of metabolizing omega-6 and omega-3 polyunsaturated fatty acids (n-6 and n-3 PUFAs). omega-6 91-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). doh 239-242 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 26002735-1 2015 Various members of the cytochrome P450 (CYP) superfamily have the capacity of metabolizing omega-6 and omega-3 polyunsaturated fatty acids (n-6 and n-3 PUFAs). omega-3 polyunsaturated fatty acids 103-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 26394652-1 2015 We have shown that the in vitro hepatic microsomal metabolism of pyranocoumarin compound decursinol angelate (DA) to decursinol (DOH) exclusively requires cytochrome P450 (CYP) enzymes, whereas the conversion of its isomer decursin (D) to DOH can be mediated by CYP and esterase(s). doh 239-242 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 26002735-1 2015 Various members of the cytochrome P450 (CYP) superfamily have the capacity of metabolizing omega-6 and omega-3 polyunsaturated fatty acids (n-6 and n-3 PUFAs). omega-3 polyunsaturated fatty acids 103-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 26002735-3 2015 The individual CYP enzymes differ in their substrate specificities as well as regio- and stereoselectivities and thus produce distinct sets of epoxy and/or hydroxy metabolites, collectively termed CYP eicosanoids. Eicosanoids 201-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 26002735-3 2015 The individual CYP enzymes differ in their substrate specificities as well as regio- and stereoselectivities and thus produce distinct sets of epoxy and/or hydroxy metabolites, collectively termed CYP eicosanoids. Eicosanoids 201-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-200 25864949-7 2015 The cytochrome P450 (CYP)-specific bioactivity of the liver microsomal film on the catalytically superior, stable HPG surface was confirmed by monitoring the electrocatalytic conversion of testosterone to 6beta-hydroxytestosterone and its inhibition by the CYP-specific ketoconazole inhibitor. Testosterone 189-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 26002735-4 2015 Nutrition has a major impact on the endogenous CYP-eicosanoid profile. Eicosanoids 51-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 25864949-7 2015 The cytochrome P450 (CYP)-specific bioactivity of the liver microsomal film on the catalytically superior, stable HPG surface was confirmed by monitoring the electrocatalytic conversion of testosterone to 6beta-hydroxytestosterone and its inhibition by the CYP-specific ketoconazole inhibitor. Testosterone 189-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 26002735-6 2015 In general, CYP eicosanoids are formed as second messengers of numerous hormones, growth factors and cytokines regulating cardiovascular and renal function, and a variety of other physiological processes. Eicosanoids 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 25864949-7 2015 The cytochrome P450 (CYP)-specific bioactivity of the liver microsomal film on the catalytically superior, stable HPG surface was confirmed by monitoring the electrocatalytic conversion of testosterone to 6beta-hydroxytestosterone and its inhibition by the CYP-specific ketoconazole inhibitor. 6 beta-hydroxytestosterone 205-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 25864949-7 2015 The cytochrome P450 (CYP)-specific bioactivity of the liver microsomal film on the catalytically superior, stable HPG surface was confirmed by monitoring the electrocatalytic conversion of testosterone to 6beta-hydroxytestosterone and its inhibition by the CYP-specific ketoconazole inhibitor. 6 beta-hydroxytestosterone 205-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 25864949-7 2015 The cytochrome P450 (CYP)-specific bioactivity of the liver microsomal film on the catalytically superior, stable HPG surface was confirmed by monitoring the electrocatalytic conversion of testosterone to 6beta-hydroxytestosterone and its inhibition by the CYP-specific ketoconazole inhibitor. Ketoconazole 270-282 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 25864949-7 2015 The cytochrome P450 (CYP)-specific bioactivity of the liver microsomal film on the catalytically superior, stable HPG surface was confirmed by monitoring the electrocatalytic conversion of testosterone to 6beta-hydroxytestosterone and its inhibition by the CYP-specific ketoconazole inhibitor. Ketoconazole 270-282 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 26002735-7 2015 Imbalances in the formation of individual CYP eicosanoids are linked to the development of hypertension, myocardial infarction, maladaptive cardiac hypertrophy, acute kidney injury, stroke and inflammatory disorders. Eicosanoids 46-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 25336106-6 2015 Among the cells tested, HepG2 cells were highly responsive to CYP inducers, such as 3-methylcholanthrene for CYP1A2 and phenobarbital for CYP2B6 and CYP3A4. Methylcholanthrene 84-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 25336106-6 2015 Among the cells tested, HepG2 cells were highly responsive to CYP inducers, such as 3-methylcholanthrene for CYP1A2 and phenobarbital for CYP2B6 and CYP3A4. Phenobarbital 120-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 26357656-2 2015 In order to investigate the effects of MGCD0103 on the metabolic capacity of cytochrome P450 (CYP) enzymes, a cocktail method was employed to evaluate the activities of human CYP2B1, CYP1A2, CYP2C11, CYP2D6, CYP3A4, and CYP2C9. mocetinostat 39-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 26357656-2 2015 In order to investigate the effects of MGCD0103 on the metabolic capacity of cytochrome P450 (CYP) enzymes, a cocktail method was employed to evaluate the activities of human CYP2B1, CYP1A2, CYP2C11, CYP2D6, CYP3A4, and CYP2C9. mocetinostat 39-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 25417047-1 2015 PURPOSE: Imatinib is a substrate of drug transporters and metabolizing enzymes, including members of the cytochrome P450 (CYP) system. Imatinib Mesylate 9-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 25417047-1 2015 PURPOSE: Imatinib is a substrate of drug transporters and metabolizing enzymes, including members of the cytochrome P450 (CYP) system. Imatinib Mesylate 9-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 26467069-3 2015 In particular, flavonoids could interfere with statins" bioavailability through different mechanisms, such as competition with cytochrome P450 (CYP) enzymes, esterases, uridine diphosphate glucuronosyltransferases and transporters (P-glycoprotein, multi-drug resistance-associated proteins, organic anion transporting polypeptides, breast cancer-resistance protein and monocarboxylate transporters). Flavonoids 15-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-142 26467069-3 2015 In particular, flavonoids could interfere with statins" bioavailability through different mechanisms, such as competition with cytochrome P450 (CYP) enzymes, esterases, uridine diphosphate glucuronosyltransferases and transporters (P-glycoprotein, multi-drug resistance-associated proteins, organic anion transporting polypeptides, breast cancer-resistance protein and monocarboxylate transporters). Flavonoids 15-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-147 26526837-0 2015 Cytochrome P450-Mediated Estrogen Metabolites and Autoimmunity: Relationship and Link to Free Radicals. Free Radicals 89-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 25200585-10 2015 Clozapine and risperidone concentration-to-dose ratios are provided as two examples of this approach of how to integrate CYP genotyping and TDM in psychiatry. Clozapine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 26526837-4 2015 Here we report that cytochrome P450-mediated estrogen metabolites produce high ROS concentrations that can result in DNA damage. ros 79-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 26639695-3 2015 The non-responsiveness to clopidogrel in cardiac patients of different populations is due to genetic variations in the cytochrome P450 (CYP) gene [2]. Clopidogrel 26-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-134 26639695-3 2015 The non-responsiveness to clopidogrel in cardiac patients of different populations is due to genetic variations in the cytochrome P450 (CYP) gene [2]. Clopidogrel 26-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-139 26639695-6 2015 Such mutations in this variant allele are responsible for the inability of the CYP enzyme to convert clopidogrel into its active metabolite, which may result in the increased risk of death, heart attack or stroke among patients who have undergone percutaneous coronary intervention (PCI) [5]. Clopidogrel 101-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 25427230-0 2015 Cytochrome P450 eicosanoids in hypertension and renal disease. Eicosanoids 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 25200585-10 2015 Clozapine and risperidone concentration-to-dose ratios are provided as two examples of this approach of how to integrate CYP genotyping and TDM in psychiatry. Risperidone 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 25285405-2 2015 Palmatine (10 muM) was metabolized using suspensions of human hepatocytes and human recombinant cytochrome P450 (CYP) enzymes. palmatine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-111 25285405-2 2015 Palmatine (10 muM) was metabolized using suspensions of human hepatocytes and human recombinant cytochrome P450 (CYP) enzymes. palmatine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 25285405-3 2015 Our analyses using electrospray ionization-quadrupole time-of-flight mass spectrometry revealed that palmatine was relatively resistant to the metabolic activity of human hepatocytes and recombinant CYP enzymes. palmatine 101-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 199-202 25666703-8 2015 Dual antiplatelet therapy with ASA and clopidogrel increases the risk of gastrointestinal bleeding in patients with acute coronary syndrome in whom concomitant treatment with a proton-pump inhibitor (PPI) was less effective owing to the interaction of clopidogrel and PPI with the same hepatic cytochrome P-450. Aspirin 31-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-310 25454080-8 2015 Lastly, different CYP families were induced in RL-14 cells using 2,3,7,8-tetrachlorodibenzo-p-dioxin and fenofibrate at mRNA and protein levels. Polychlorinated Dibenzodioxins 65-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 25454080-8 2015 Lastly, different CYP families were induced in RL-14 cells using 2,3,7,8-tetrachlorodibenzo-p-dioxin and fenofibrate at mRNA and protein levels. Fenofibrate 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 25666703-8 2015 Dual antiplatelet therapy with ASA and clopidogrel increases the risk of gastrointestinal bleeding in patients with acute coronary syndrome in whom concomitant treatment with a proton-pump inhibitor (PPI) was less effective owing to the interaction of clopidogrel and PPI with the same hepatic cytochrome P-450. Clopidogrel 39-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-310 25518709-5 2014 METHODS: Stable isotope 13C-labelled primaquine and its two enantiomers were incubated with recombinant cytochrome-P450 supersomes containing CYP2D6 under optimized conditions. 13c 24-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 26514046-6 2015 Some antidepressants, such as paroxetine and fluvoxamine, are strong inhibitors of the CYP system. Paroxetine 30-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 26514046-6 2015 Some antidepressants, such as paroxetine and fluvoxamine, are strong inhibitors of the CYP system. Fluvoxamine 45-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 25579709-7 2015 All our subjects received rifabutin instead of rifampin in the anti-TB treatment regime as rifabutin is a less-potent inducer of cytochrome P-450. Rifabutin 91-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 25518709-5 2014 METHODS: Stable isotope 13C-labelled primaquine and its two enantiomers were incubated with recombinant cytochrome-P450 supersomes containing CYP2D6 under optimized conditions. Primaquine 37-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 25426071-1 2014 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid through cytochrome P450 (CYP) epoxygenases have many biological functions. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 26579418-1 2014 Cytochrome P450 (CYP) enzymes metabolize numerous endogenous substrates, such as retinoids, androgens, estrogens and vitamin D, that can modulate important cellular processes, including proliferation, differentiation and apoptosis. Retinoids 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26579418-1 2014 Cytochrome P450 (CYP) enzymes metabolize numerous endogenous substrates, such as retinoids, androgens, estrogens and vitamin D, that can modulate important cellular processes, including proliferation, differentiation and apoptosis. Retinoids 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 26579418-1 2014 Cytochrome P450 (CYP) enzymes metabolize numerous endogenous substrates, such as retinoids, androgens, estrogens and vitamin D, that can modulate important cellular processes, including proliferation, differentiation and apoptosis. Vitamin D 117-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26579418-1 2014 Cytochrome P450 (CYP) enzymes metabolize numerous endogenous substrates, such as retinoids, androgens, estrogens and vitamin D, that can modulate important cellular processes, including proliferation, differentiation and apoptosis. Vitamin D 117-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 25047139-1 2014 WHAT IS KNOWN AND OBJECTIVE: Navitoclax, a first-in-class small molecule Bcl-2 family inhibitor, is metabolized in vitro by the hepatic microsomal cytochrome P450 (CYP) enzymes CYP3A4. navitoclax 29-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-162 25047139-1 2014 WHAT IS KNOWN AND OBJECTIVE: Navitoclax, a first-in-class small molecule Bcl-2 family inhibitor, is metabolized in vitro by the hepatic microsomal cytochrome P450 (CYP) enzymes CYP3A4. navitoclax 29-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-167 25520592-2 2014 Herein we report a systematic investigation of the impacts of gold nanoparticles (AuNPs) on five major CYP isozymes under in vitro incubations of human liver microsomes (HLMs) with tannic acid (TA)-stabilized AuNPs in the size range of 5 to 100 nm. Tannins 181-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 25148377-2 2014 This suggestion was predicated on observations of highly attenuated morphine antinociception in rodents after intracerebroventricular injection of fluconazole or carrying a neuron-specific deletion of the cytochrome P450 reductase. Morphine 68-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-220 25148377-2 2014 This suggestion was predicated on observations of highly attenuated morphine antinociception in rodents after intracerebroventricular injection of fluconazole or carrying a neuron-specific deletion of the cytochrome P450 reductase. Fluconazole 147-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-220 25148377-4 2014 Although CYP inhibitors such as fluconazole are unlikely to attenuate remifentanil analgesia in humans, extrapolation of the findings to other opioids is premature because differences among opioid effects, such as ligand-selective biased signaling at opioid receptors, leave the possibility that CYP-dependent opioid signaling in the brain might be limited to morphine and may not extend to remifentanil. Fluconazole 32-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 25426071-1 2014 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid through cytochrome P450 (CYP) epoxygenases have many biological functions. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-93 25426071-1 2014 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid through cytochrome P450 (CYP) epoxygenases have many biological functions. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 25426071-1 2014 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid through cytochrome P450 (CYP) epoxygenases have many biological functions. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-93 25426071-1 2014 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid through cytochrome P450 (CYP) epoxygenases have many biological functions. Arachidonic Acid 48-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 25426071-1 2014 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid through cytochrome P450 (CYP) epoxygenases have many biological functions. Arachidonic Acid 48-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-93 24882266-1 2014 Beyond their contribution to the metabolism of xenobiotics, cytochrome P450 (CYP) epoxygenases are actively involved in the metabolism of endogenous substances, like arachidonic acid (AA). Arachidonic Acid 166-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 24803100-1 2014 AIMS: Variability in responsiveness to clopidogrel is a clinical problem in secondary prevention after cerebral ischaemia which has been suggested to be linked to competitive metabolization of clopidogrel and cytochrome P450 (CYP) 3A4-oxidated statins such as simvastatin. Clopidogrel 39-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-224 24803100-1 2014 AIMS: Variability in responsiveness to clopidogrel is a clinical problem in secondary prevention after cerebral ischaemia which has been suggested to be linked to competitive metabolization of clopidogrel and cytochrome P450 (CYP) 3A4-oxidated statins such as simvastatin. Clopidogrel 39-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-229 24803100-1 2014 AIMS: Variability in responsiveness to clopidogrel is a clinical problem in secondary prevention after cerebral ischaemia which has been suggested to be linked to competitive metabolization of clopidogrel and cytochrome P450 (CYP) 3A4-oxidated statins such as simvastatin. Simvastatin 260-271 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-224 24803100-1 2014 AIMS: Variability in responsiveness to clopidogrel is a clinical problem in secondary prevention after cerebral ischaemia which has been suggested to be linked to competitive metabolization of clopidogrel and cytochrome P450 (CYP) 3A4-oxidated statins such as simvastatin. Simvastatin 260-271 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-229 24803100-10 2014 CONCLUSIONS: Our data indicate that treatment with CYP 3A4-metabolized simvastatin does not jeopardize clopidogrel-mediated inhibition of platelet aggregation. Simvastatin 71-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 25363545-0 2014 Cytochrome P450 single nucleotide polymorphisms in an indigenous Tanzanian population: a concern about the metabolism of artemisinin-based combinations. artemisinin 121-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24944083-0 2014 Identification of human cytochrome P450 isozymes involved in the metabolism of naftopidil enantiomers in vitro. naftopidil 79-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 24882266-1 2014 Beyond their contribution to the metabolism of xenobiotics, cytochrome P450 (CYP) epoxygenases are actively involved in the metabolism of endogenous substances, like arachidonic acid (AA). Arachidonic Acid 166-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 24882266-2 2014 The main human CYP epoxygenases, i.e. CYP2C8, CYP2C9, CYP2C19 and CYP2J2, convert AA to four regioisomer epoxyeicosatrienoic acids (EETs). epoxyeicosatrienoic acids 105-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 24882266-2 2014 The main human CYP epoxygenases, i.e. CYP2C8, CYP2C9, CYP2C19 and CYP2J2, convert AA to four regioisomer epoxyeicosatrienoic acids (EETs). eets 132-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 24681287-0 2014 Impact of 17-alpha-hydroxyprogesterone caproate on cytochrome P450s in primary cultures of human hepatocytes. 17 alpha-Hydroxyprogesterone Caproate 10-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-66 25285807-4 2014 The oxidation reactions mediated by CYP450 are known to occur by either a single electron transfer (SET) or a hydrogen atom transfer (HAT) mechanism. Hydrogen 110-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-42 25238648-0 2014 Effect of axial ligand, spin state, and hydrogen bonding on the inner-sphere reorganization energies of functional models of cytochrome P450. Hydrogen 40-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-140 25238648-8 2014 The results suggest that while the hydrogen bonding to the thiolate in the 5C HS thiolate bound active site of cytochrome P450 (cyp450) shifts the potential up, resulting in a negative DeltaG, it also increases lambda resulting in an overall low barrier for the electron transfer process. Hydrogen 35-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-126 25238648-8 2014 The results suggest that while the hydrogen bonding to the thiolate in the 5C HS thiolate bound active site of cytochrome P450 (cyp450) shifts the potential up, resulting in a negative DeltaG, it also increases lambda resulting in an overall low barrier for the electron transfer process. Hydrogen 35-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-134 25238648-8 2014 The results suggest that while the hydrogen bonding to the thiolate in the 5C HS thiolate bound active site of cytochrome P450 (cyp450) shifts the potential up, resulting in a negative DeltaG, it also increases lambda resulting in an overall low barrier for the electron transfer process. thiolate 59-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-126 25238648-8 2014 The results suggest that while the hydrogen bonding to the thiolate in the 5C HS thiolate bound active site of cytochrome P450 (cyp450) shifts the potential up, resulting in a negative DeltaG, it also increases lambda resulting in an overall low barrier for the electron transfer process. thiolate 59-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-134 25238648-8 2014 The results suggest that while the hydrogen bonding to the thiolate in the 5C HS thiolate bound active site of cytochrome P450 (cyp450) shifts the potential up, resulting in a negative DeltaG, it also increases lambda resulting in an overall low barrier for the electron transfer process. thiolate 81-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-126 25238648-8 2014 The results suggest that while the hydrogen bonding to the thiolate in the 5C HS thiolate bound active site of cytochrome P450 (cyp450) shifts the potential up, resulting in a negative DeltaG, it also increases lambda resulting in an overall low barrier for the electron transfer process. thiolate 81-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-134 24681287-1 2014 OBJECTIVE: The aim of this study was to examine the effects of 17-alpha-hydroxyprogesterone caproate (17OHP-C) on the activity and expression of several common hepatic cytochrome P450 (CYP) enzymes. 17 alpha-Hydroxyprogesterone Caproate 63-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-183 24681287-1 2014 OBJECTIVE: The aim of this study was to examine the effects of 17-alpha-hydroxyprogesterone caproate (17OHP-C) on the activity and expression of several common hepatic cytochrome P450 (CYP) enzymes. 17 alpha-Hydroxyprogesterone Caproate 63-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-188 24681287-1 2014 OBJECTIVE: The aim of this study was to examine the effects of 17-alpha-hydroxyprogesterone caproate (17OHP-C) on the activity and expression of several common hepatic cytochrome P450 (CYP) enzymes. 17 alpha-Hydroxyprogesterone Caproate 102-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-183 24681287-1 2014 OBJECTIVE: The aim of this study was to examine the effects of 17-alpha-hydroxyprogesterone caproate (17OHP-C) on the activity and expression of several common hepatic cytochrome P450 (CYP) enzymes. 17 alpha-Hydroxyprogesterone Caproate 102-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-188 25047407-2 2014 This open-label, single-centre, 2-period study evaluated the effect of armodafinil, a moderate inducer of cytochrome-P450 (CYP) isoenzyme CYP3A4, on the pharmacokinetics and safety of ziprasidone, an atypical antipsychotic used to treat bipolar I disorder and metabolized in part by CYP3A4. Modafinil 71-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 25047407-2 2014 This open-label, single-centre, 2-period study evaluated the effect of armodafinil, a moderate inducer of cytochrome-P450 (CYP) isoenzyme CYP3A4, on the pharmacokinetics and safety of ziprasidone, an atypical antipsychotic used to treat bipolar I disorder and metabolized in part by CYP3A4. Modafinil 71-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 24535830-2 2014 In vitro assays based on the use of human liver microsomes and recombinant cytochrome P450 isoforms were developed and applied to characterize the phase I metabolic profile of the prohibited agent stanozolol, both in the absence and in the presence of substances (ketoconazole, itraconazole, miconazole, cimetidine, ranitidine, and nefazodone) not included in the World Anti-Doping Agency (WADA) list of prohibited substances and methods and frequently administered to athletes. Stanozolol 197-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 24987833-4 2014 Components of cancer inflammation include chemokines, prostaglandins, and cytokines, and these have been shown to downregulate cytochrome P450 (CYP) enzyme activity. Prostaglandins 54-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-142 24987833-4 2014 Components of cancer inflammation include chemokines, prostaglandins, and cytokines, and these have been shown to downregulate cytochrome P450 (CYP) enzyme activity. Prostaglandins 54-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-147 25048408-0 2014 Impact of cytochrome P450 inducers with or without inhibitors on the serum clobazam level in patients with antiepileptic polypharmacy. Clobazam 75-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 25048408-1 2014 PURPOSE: The aim of this study was to evaluate the effect of cytochrome P450 (CYP) inducers/inhibitors on the pharmacokinetics of clobazam (CLB) in patients receiving antiepileptic polypharmacy. Clobazam 130-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 25048408-1 2014 PURPOSE: The aim of this study was to evaluate the effect of cytochrome P450 (CYP) inducers/inhibitors on the pharmacokinetics of clobazam (CLB) in patients receiving antiepileptic polypharmacy. Clobazam 130-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 25048408-1 2014 PURPOSE: The aim of this study was to evaluate the effect of cytochrome P450 (CYP) inducers/inhibitors on the pharmacokinetics of clobazam (CLB) in patients receiving antiepileptic polypharmacy. Clobazam 140-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 25048408-1 2014 PURPOSE: The aim of this study was to evaluate the effect of cytochrome P450 (CYP) inducers/inhibitors on the pharmacokinetics of clobazam (CLB) in patients receiving antiepileptic polypharmacy. Clobazam 140-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 24128439-2 2014 We examined whether DNA methylation levels of Cytochrome P450 (CYP) enzymes (CYP2R1, CYP24A1, CYP27A1, and CYP27B1) are potential biomarkers predicting vitamin D response variation. Vitamin D 152-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 24128439-2 2014 We examined whether DNA methylation levels of Cytochrome P450 (CYP) enzymes (CYP2R1, CYP24A1, CYP27A1, and CYP27B1) are potential biomarkers predicting vitamin D response variation. Vitamin D 152-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. Arachidonic Acid 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 25151892-0 2014 Urinary CYP eicosanoid excretion correlates with glomerular filtration in African-Americans with chronic kidney disease. Eicosanoids 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-11 25068656-3 2014 Mice, unlike humans, have the ability to detoxify hydrophobic bile salts by cytochrome P450-mediated (re)hydroxylation and thus have a less toxic bile salt pool. Bile Acids and Salts 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 25068656-3 2014 Mice, unlike humans, have the ability to detoxify hydrophobic bile salts by cytochrome P450-mediated (re)hydroxylation and thus have a less toxic bile salt pool. Bile Acids and Salts 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 25484616-3 2014 Experimental animal and human data indicate that TCE metabolism occurs through two major pathways: cytochrome P450 (CYP)-dependent oxidation and glutathione (GSH) conjugation catalyzed by GSH S-transferases (GSTs). Trichloroethylene 49-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-114 25484616-3 2014 Experimental animal and human data indicate that TCE metabolism occurs through two major pathways: cytochrome P450 (CYP)-dependent oxidation and glutathione (GSH) conjugation catalyzed by GSH S-transferases (GSTs). Trichloroethylene 49-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. Arachidonic Acid 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 103-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 103-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 136-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 136-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. epoxyeicosatrienoic acids 149-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. epoxyeicosatrienoic acids 149-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. eets 176-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 25151892-1 2014 Previous studies have indicated that cytochrome P450 (CYP) metabolites of arachidonic acid (AA), i.e., 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), play an important role in the regulation of renal tubular and vascular function. eets 176-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 25061110-9 2014 Because CYP3A4/5/7 represent up to 40% of total cytochrome P450 in the liver, these data indicate that these enzymes are the major source of H2O2 in human liver microsomes. Hydrogen Peroxide 141-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 25061110-0 2014 Human recombinant cytochrome P450 enzymes display distinct hydrogen peroxide generating activities during substrate independent NADPH oxidase reactions. Hydrogen Peroxide 59-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 25408661-3 2014 Ifosfamide is required to be activated by hepatic cytochrome P450 (CYP), especially the 3A4 subtype, leading to 4-hydroxy-ifosfamide. Ifosfamide 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 25061110-3 2014 In the presence of redox active transition metals, H2O2 can form highly toxic hydroxyl radicals and, depending on the "oxidase" activity of individual cytochrome P450 isoenzymes, this can compromise cellular functioning and contribute to tissue injury. Hydrogen Peroxide 51-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-166 25061110-6 2014 Significantly greater activity was detected in preparations containing individual cytochrome P450s coexpressed with CPR (from 6.0 nmol H2O2/min/nmol P450 to 0.2 nmol/min/nmol P450); CYP1A1 was the most active, followed by CYP2D6, CYP3A4, CYP2E1, CYP4A11, CYP1A2, and CYP2C subfamily enzymes. Hydrogen Peroxide 135-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 25061110-7 2014 H2O2 generating activity of the cytochrome P450s was independent of the ratio of CYP/CPR. Hydrogen Peroxide 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 24929095-3 2014 In addition, normal CYP activity may produce reactive oxygen species (ROS) that contribute to cell damage through oxidative mechanisms. Reactive Oxygen Species 45-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-23 24929095-3 2014 In addition, normal CYP activity may produce reactive oxygen species (ROS) that contribute to cell damage through oxidative mechanisms. Reactive Oxygen Species 70-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-23 24929095-4 2014 CYP2E1 is a CYP isoform that can generate ROS leading to cytotoxicity in multiple tissue types. Reactive Oxygen Species 42-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 25408661-3 2014 Ifosfamide is required to be activated by hepatic cytochrome P450 (CYP), especially the 3A4 subtype, leading to 4-hydroxy-ifosfamide. Ifosfamide 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 25408661-3 2014 Ifosfamide is required to be activated by hepatic cytochrome P450 (CYP), especially the 3A4 subtype, leading to 4-hydroxy-ifosfamide. 4-hydroxyifosfamide 112-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 25408661-3 2014 Ifosfamide is required to be activated by hepatic cytochrome P450 (CYP), especially the 3A4 subtype, leading to 4-hydroxy-ifosfamide. 4-hydroxyifosfamide 112-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 24949552-10 2014 In this study, GSTM1 which is known to be highly polymorphic within the human population, but so far could not be linked to toxicity in acetaminophen-poisoned patients, is suggested to cause interindividual variability in acetaminophen-metabolism, dependent on the individual"s gene expression-responses of CYP-enzymes. Acetaminophen 222-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 307-310 25408844-2 2014 Among substrates for CYP450 is the tryptophan metabolite skatole (3-methylindole), one of the major contributors to the off-odour associated with boar-tainted meat. Tryptophan 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-27 25408844-2 2014 Among substrates for CYP450 is the tryptophan metabolite skatole (3-methylindole), one of the major contributors to the off-odour associated with boar-tainted meat. Skatole 57-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-27 25408844-2 2014 Among substrates for CYP450 is the tryptophan metabolite skatole (3-methylindole), one of the major contributors to the off-odour associated with boar-tainted meat. Skatole 66-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-27 24926821-3 2014 The level of the active metabolite of vitamin D, 1alpha,25-dihydroxyvitamin D3 (1,25D), is controlled in part by VDR-dependent induction of cytochrome P450, family 24, subfamily 1, polypeptide1 (CYP24A1), which metabolizes 1,25D to an inactive form. Vitamin D 38-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-179 24973543-2 2014 Ritonavir pharmacokinetics are complicated by inhibition, induction and pharmacogenetics of cytochrome P450 (CYP) enzymes mediating its clearance. Ritonavir 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 24973543-2 2014 Ritonavir pharmacokinetics are complicated by inhibition, induction and pharmacogenetics of cytochrome P450 (CYP) enzymes mediating its clearance. Ritonavir 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-112 24548191-1 2014 AIMS: To examine whether initiation of fibrates or statins in sulfonylurea users is associated with hypoglycaemia, and examine in vitro inhibition of cytochrome P450 (CYP) enzymes by statins, fenofibrate and glipizide. Fenofibrate 192-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-170 24548191-1 2014 AIMS: To examine whether initiation of fibrates or statins in sulfonylurea users is associated with hypoglycaemia, and examine in vitro inhibition of cytochrome P450 (CYP) enzymes by statins, fenofibrate and glipizide. Glipizide 208-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-170 24548191-3 2014 We also characterized the in vitro inhibition of CYP enzymes by statins, fenofibrate and glipizide using fluorometric CYP450 inhibition assays, and estimated area under the concentration-time curve ratios (AUCRs) for the drug pairs. Fenofibrate 73-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 24548191-3 2014 We also characterized the in vitro inhibition of CYP enzymes by statins, fenofibrate and glipizide using fluorometric CYP450 inhibition assays, and estimated area under the concentration-time curve ratios (AUCRs) for the drug pairs. Glipizide 89-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 24548191-7 2014 The predicted CYP-based AUCRs for fenofibrate-glyburide and gemfibrozil-glyburide interactions were only 1.09 and 1.04, suggesting that CYP inhibition is unlikely to explain such an interaction. fenofibrate-glyburide 34-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 24548191-7 2014 The predicted CYP-based AUCRs for fenofibrate-glyburide and gemfibrozil-glyburide interactions were only 1.09 and 1.04, suggesting that CYP inhibition is unlikely to explain such an interaction. gemfibrozil-glyburide 60-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 24926821-3 2014 The level of the active metabolite of vitamin D, 1alpha,25-dihydroxyvitamin D3 (1,25D), is controlled in part by VDR-dependent induction of cytochrome P450, family 24, subfamily 1, polypeptide1 (CYP24A1), which metabolizes 1,25D to an inactive form. Calcitriol 49-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-179 25114852-1 2014 Biotransformation of tamoxifen to the potent antiestrogen endoxifen is performed by cytochrome P450 (CYP) enzymes, in particular the CYP2D6 isoform. Tamoxifen 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 24590553-3 2014 The present study demonstrates that such control is possible by using wild type or mutant forms of the monooxygenase cytochrome P450 BM3 as catalysts in the oxidative hydroxylation of methylcyclohexane and seven other monosubstituted cyclohexane derivatives. methylcyclohexane 184-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-132 24590553-3 2014 The present study demonstrates that such control is possible by using wild type or mutant forms of the monooxygenase cytochrome P450 BM3 as catalysts in the oxidative hydroxylation of methylcyclohexane and seven other monosubstituted cyclohexane derivatives. cyclohexane derivatives 234-257 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-132 24722393-8 2014 Minimally invasive one- and three-point (at 2, 3, and 6 h) DBS-sampling methods were found to reliably reflect CYP and P-gp activities at each session. dbs 59-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-114 24981927-4 2014 Cytochrome P450 (CYP) enzymes activate the xenobiotic making it more reactive, while the Glutathione S-transferases (GST) enzymes conjugate the reduced glutathione with electrophilic compounds, facilitating the toxic products excretion. Glutathione 152-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24981927-4 2014 Cytochrome P450 (CYP) enzymes activate the xenobiotic making it more reactive, while the Glutathione S-transferases (GST) enzymes conjugate the reduced glutathione with electrophilic compounds, facilitating the toxic products excretion. Glutathione 152-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 25123135-0 2014 Mammalian cytochrome P450-dependent metabolism of polychlorinated dibenzo-p-dioxins and coplanar polychlorinated biphenyls. Polychlorinated Dibenzodioxins 50-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 25123135-0 2014 Mammalian cytochrome P450-dependent metabolism of polychlorinated dibenzo-p-dioxins and coplanar polychlorinated biphenyls. Polychlorinated Biphenyls 97-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 25123135-3 2014 A number of human CYP isoforms belonging to the CYP1 and CYP2 families showed remarkable activities toward low-chlorinated PCDDs. Polychlorinated Dibenzodioxins 123-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 25114852-1 2014 Biotransformation of tamoxifen to the potent antiestrogen endoxifen is performed by cytochrome P450 (CYP) enzymes, in particular the CYP2D6 isoform. Tamoxifen 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 25114852-1 2014 Biotransformation of tamoxifen to the potent antiestrogen endoxifen is performed by cytochrome P450 (CYP) enzymes, in particular the CYP2D6 isoform. 4-hydroxy-N-desmethyltamoxifen 58-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 25114852-1 2014 Biotransformation of tamoxifen to the potent antiestrogen endoxifen is performed by cytochrome P450 (CYP) enzymes, in particular the CYP2D6 isoform. 4-hydroxy-N-desmethyltamoxifen 58-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 24889073-0 2014 Cytochrome P450 3A-mediated metabolism of the topoisomerase I inhibitor 9-aminocamptothecin: impact on cancer therapy. 9-aminocamptothecin 72-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24194261-3 2014 Here, to investigate the inhibitory effects of PL on the activities of CYP isoforms, CYP inhibition assays were conducted using a cocktail of probe substrates in pooled human liver microsome (HLMs) and human recombinant cDNA-expressed CYP. piperlonguminine 47-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 25056785-2 2014 The aim of the present work was to evaluate the impact of arecoline on human hepatic cytochrome P450 (CYP) enzymes in vitro and rat hepatic CYP enzymes, as well as the hepatic oxidative stress and liver injury of rats in vivo. Arecoline 58-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 24816159-7 2014 The specific cytochrome P450 (CYP) enzymes responsible for m-nisoldipine metabolites were identified using chemical inhibition and cDNA expressed CYP enzymes. m-Nisoldipine 59-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 24816159-7 2014 The specific cytochrome P450 (CYP) enzymes responsible for m-nisoldipine metabolites were identified using chemical inhibition and cDNA expressed CYP enzymes. m-Nisoldipine 59-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 24816159-7 2014 The specific cytochrome P450 (CYP) enzymes responsible for m-nisoldipine metabolites were identified using chemical inhibition and cDNA expressed CYP enzymes. m-Nisoldipine 59-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 25056785-2 2014 The aim of the present work was to evaluate the impact of arecoline on human hepatic cytochrome P450 (CYP) enzymes in vitro and rat hepatic CYP enzymes, as well as the hepatic oxidative stress and liver injury of rats in vivo. Arecoline 58-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 24839071-1 2014 The purpose of this study is to characterize the involvement of hepato-biliary transport and cytochrome-P450 (CYP)-mediated metabolism in the disposition of glyburide and predict its pharmacokinetic variability due to drug interactions and genetic variations. Glyburide 157-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-108 24575896-0 2014 Contribution of cytochrome P450 and UDT-glucuronosyltransferase to the metabolism of drugs containing carboxylic acid groups: risk assessment of acylglucuronides using human hepatocytes. Carboxylic Acids 102-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 24575896-0 2014 Contribution of cytochrome P450 and UDT-glucuronosyltransferase to the metabolism of drugs containing carboxylic acid groups: risk assessment of acylglucuronides using human hepatocytes. acylglucuronides 145-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 24575896-2 2014 In order to evaluate the inhibition activity of 1-aminobenzotriazole (ABT) and (-)-borneol (borneol) against cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT), the substrates of these metabolic enzymes were incubated with ABT and borneol in human hepatocytes. 1-aminobenzotriazole 48-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 24575896-2 2014 In order to evaluate the inhibition activity of 1-aminobenzotriazole (ABT) and (-)-borneol (borneol) against cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT), the substrates of these metabolic enzymes were incubated with ABT and borneol in human hepatocytes. 1-aminobenzotriazole 48-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 24575896-2 2014 In order to evaluate the inhibition activity of 1-aminobenzotriazole (ABT) and (-)-borneol (borneol) against cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT), the substrates of these metabolic enzymes were incubated with ABT and borneol in human hepatocytes. 1-aminobenzotriazole 70-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 24710199-2 2014 The tutorial instructs the reader how to construct the VB diagrams and how to estimate HAT barriers from raw data, starting with the simplest reaction H + H2 and going all the way to HAT in the enzyme cytochrome P450. Hydrogen 155-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-216 24575896-2 2014 In order to evaluate the inhibition activity of 1-aminobenzotriazole (ABT) and (-)-borneol (borneol) against cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT), the substrates of these metabolic enzymes were incubated with ABT and borneol in human hepatocytes. 1-aminobenzotriazole 70-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 24575896-2 2014 In order to evaluate the inhibition activity of 1-aminobenzotriazole (ABT) and (-)-borneol (borneol) against cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT), the substrates of these metabolic enzymes were incubated with ABT and borneol in human hepatocytes. isoborneol 79-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 24575896-2 2014 In order to evaluate the inhibition activity of 1-aminobenzotriazole (ABT) and (-)-borneol (borneol) against cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT), the substrates of these metabolic enzymes were incubated with ABT and borneol in human hepatocytes. isoborneol 79-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 24575896-3 2014 We found that 3 mM ABT and 300 muM borneol were the most suitable experimental levels to specifically inhibit CYP and UGT. 1-aminobenzotriazole 19-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 24575896-3 2014 We found that 3 mM ABT and 300 muM borneol were the most suitable experimental levels to specifically inhibit CYP and UGT. isoborneol 35-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 25024195-1 2014 Prostaglandins derived from the cyclooxygenase (COX) pathway and epoxyeicosatrienoic acids (EETs) from the cytochrome P450/soluble epoxide hydrolase (sEH) pathway are important eicosanoids that regulate angiogenesis and tumorigenesis. epoxyeicosatrienoic acids 65-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-148 25024195-1 2014 Prostaglandins derived from the cyclooxygenase (COX) pathway and epoxyeicosatrienoic acids (EETs) from the cytochrome P450/soluble epoxide hydrolase (sEH) pathway are important eicosanoids that regulate angiogenesis and tumorigenesis. eets 92-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-148 25024195-1 2014 Prostaglandins derived from the cyclooxygenase (COX) pathway and epoxyeicosatrienoic acids (EETs) from the cytochrome P450/soluble epoxide hydrolase (sEH) pathway are important eicosanoids that regulate angiogenesis and tumorigenesis. Eicosanoids 177-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-148 24956138-3 2014 In this work, the ionization potentials (IPs) of the S-NPC1161 (NPC1161a) hydroxylated derivatives, which are possible metabolites derived from action of endogenous cytochrome P450 (CYP450) enzymes, were calculated at the B3LYP-SCRF(PCM)/6-311++G**//B3LYP/6-31G** level in water. IPS 41-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-180 24956138-3 2014 In this work, the ionization potentials (IPs) of the S-NPC1161 (NPC1161a) hydroxylated derivatives, which are possible metabolites derived from action of endogenous cytochrome P450 (CYP450) enzymes, were calculated at the B3LYP-SCRF(PCM)/6-311++G**//B3LYP/6-31G** level in water. IPS 41-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-188 24956138-3 2014 In this work, the ionization potentials (IPs) of the S-NPC1161 (NPC1161a) hydroxylated derivatives, which are possible metabolites derived from action of endogenous cytochrome P450 (CYP450) enzymes, were calculated at the B3LYP-SCRF(PCM)/6-311++G**//B3LYP/6-31G** level in water. Water 273-278 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-180 24956138-3 2014 In this work, the ionization potentials (IPs) of the S-NPC1161 (NPC1161a) hydroxylated derivatives, which are possible metabolites derived from action of endogenous cytochrome P450 (CYP450) enzymes, were calculated at the B3LYP-SCRF(PCM)/6-311++G**//B3LYP/6-31G** level in water. Water 273-278 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-188 24956138-8 2014 Molecular orbital analysis implies that the N1" position should be the most reactive center when NPC1161 approaches the heme in CYP450. Heme 120-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-134 24839071-1 2014 The purpose of this study is to characterize the involvement of hepato-biliary transport and cytochrome-P450 (CYP)-mediated metabolism in the disposition of glyburide and predict its pharmacokinetic variability due to drug interactions and genetic variations. Glyburide 157-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 24400699-0 2014 Effects of artemisinin antimalarials on Cytochrome P450 enzymes in vitro using recombinant enzymes and human liver microsomes: potential implications for combination therapies. artemisinin 11-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 24752505-0 2014 Contribution of carboxylesterase and cytochrome P450 to the bioactivation and detoxification of isocarbophos and its enantiomers in human liver microsomes. isocarbophos 96-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 24405273-5 2014 We evaluated rose bengal as an in vitro inhibitor of cytochrome P450 (CYP) or UDP-glucuronosyltransferase (UGT) enzymes in both human liver microsomes (HLM) and cryopreserved human hepatocytes (CHHs) under both yellow light and dark conditions. Rose Bengal 13-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 24405273-5 2014 We evaluated rose bengal as an in vitro inhibitor of cytochrome P450 (CYP) or UDP-glucuronosyltransferase (UGT) enzymes in both human liver microsomes (HLM) and cryopreserved human hepatocytes (CHHs) under both yellow light and dark conditions. Rose Bengal 13-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 24762860-8 2014 CONCLUSIONS: Both loss-of-function and gain-of-function CYP polymorphisms affecting clopidogrel metabolism are associated with increased mortality among clopidogrel-treated patients after acute myocardial infarction; the specific polymorphism and the putative mechanism vary according to race. Clopidogrel 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 24762860-1 2014 BACKGROUND: Clopidogrel is recommended after acute myocardial infarction but has variable efficacy and safety, in part related to the effect of cytochrome P450 (CYP) polymorphisms on its metabolism. Clopidogrel 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-159 24875119-3 2014 Together with the recent determination of an iron(IV)hydroxide pK(a) ~ 12 in the thiolate-ligated heme enzyme cytochrome P450, this result provides insight into Nature"s ability to tune catalytic function through its choice of axial ligand. Iron 45-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 24875119-3 2014 Together with the recent determination of an iron(IV)hydroxide pK(a) ~ 12 in the thiolate-ligated heme enzyme cytochrome P450, this result provides insight into Nature"s ability to tune catalytic function through its choice of axial ligand. hydroxide ion 52-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 24875119-3 2014 Together with the recent determination of an iron(IV)hydroxide pK(a) ~ 12 in the thiolate-ligated heme enzyme cytochrome P450, this result provides insight into Nature"s ability to tune catalytic function through its choice of axial ligand. thiolate 81-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 24762860-1 2014 BACKGROUND: Clopidogrel is recommended after acute myocardial infarction but has variable efficacy and safety, in part related to the effect of cytochrome P450 (CYP) polymorphisms on its metabolism. Clopidogrel 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 24762860-8 2014 CONCLUSIONS: Both loss-of-function and gain-of-function CYP polymorphisms affecting clopidogrel metabolism are associated with increased mortality among clopidogrel-treated patients after acute myocardial infarction; the specific polymorphism and the putative mechanism vary according to race. Clopidogrel 153-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 24634501-0 2014 Dietary omega-3 fatty acids modulate the eicosanoid profile in man primarily via the CYP-epoxygenase pathway. Fatty Acids, Omega-3 8-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 23797843-3 2014 In the present study, the metabolism of PQ was evaluated by incubating the drug with pooled human hepatocytes cultured in vitro as well as with recombinant cytochrome P450 (CYP) iso- enzymes, monoamine oxidases (MAO), and flavin-containing monooxygenases (FMO). Primaquine 40-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-171 23797843-3 2014 In the present study, the metabolism of PQ was evaluated by incubating the drug with pooled human hepatocytes cultured in vitro as well as with recombinant cytochrome P450 (CYP) iso- enzymes, monoamine oxidases (MAO), and flavin-containing monooxygenases (FMO). Primaquine 40-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-176 24634501-0 2014 Dietary omega-3 fatty acids modulate the eicosanoid profile in man primarily via the CYP-epoxygenase pathway. Eicosanoids 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 24634501-1 2014 Cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA) contribute to the regulation of cardiovascular function. Arachidonic Acid 47-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24634501-1 2014 Cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA) contribute to the regulation of cardiovascular function. Arachidonic Acid 47-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 24634501-2 2014 CYP enzymes also accept EPA and DHA to yield more potent vasodilatory and potentially anti-arrhythmic metabolites, suggesting that the endogenous CYP-eicosanoid profile can be favorably shifted by dietary omega-3 fatty acids. dehydroacetic acid 32-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 24634501-2 2014 CYP enzymes also accept EPA and DHA to yield more potent vasodilatory and potentially anti-arrhythmic metabolites, suggesting that the endogenous CYP-eicosanoid profile can be favorably shifted by dietary omega-3 fatty acids. dehydroacetic acid 32-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 24634501-2 2014 CYP enzymes also accept EPA and DHA to yield more potent vasodilatory and potentially anti-arrhythmic metabolites, suggesting that the endogenous CYP-eicosanoid profile can be favorably shifted by dietary omega-3 fatty acids. Eicosanoids 150-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 24634501-2 2014 CYP enzymes also accept EPA and DHA to yield more potent vasodilatory and potentially anti-arrhythmic metabolites, suggesting that the endogenous CYP-eicosanoid profile can be favorably shifted by dietary omega-3 fatty acids. Eicosanoids 150-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 24634501-2 2014 CYP enzymes also accept EPA and DHA to yield more potent vasodilatory and potentially anti-arrhythmic metabolites, suggesting that the endogenous CYP-eicosanoid profile can be favorably shifted by dietary omega-3 fatty acids. Fatty Acids, Omega-3 205-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 24634501-2 2014 CYP enzymes also accept EPA and DHA to yield more potent vasodilatory and potentially anti-arrhythmic metabolites, suggesting that the endogenous CYP-eicosanoid profile can be favorably shifted by dietary omega-3 fatty acids. Fatty Acids, Omega-3 205-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 24634501-5 2014 The metabolite/precursor fatty acid ratios indicated that CYP epoxygenases metabolized EPA with an 8.6-fold higher efficiency and DHA with a 2.2-fold higher efficiency than AA. Fatty Acids 25-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 24634501-5 2014 The metabolite/precursor fatty acid ratios indicated that CYP epoxygenases metabolized EPA with an 8.6-fold higher efficiency and DHA with a 2.2-fold higher efficiency than AA. dehydroacetic acid 130-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 24634501-7 2014 We propose that CYP-dependent epoxy-metabolites of EPA and DHA may function as mediators of the vasodilatory and cardioprotective effects of omega-3 fatty acids and could serve as biomarkers in clinical studies investigating the cardiovascular effects of EPA/DHA supplementation. dehydroacetic acid 59-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 24634501-7 2014 We propose that CYP-dependent epoxy-metabolites of EPA and DHA may function as mediators of the vasodilatory and cardioprotective effects of omega-3 fatty acids and could serve as biomarkers in clinical studies investigating the cardiovascular effects of EPA/DHA supplementation. Fatty Acids, Omega-3 141-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 24634501-7 2014 We propose that CYP-dependent epoxy-metabolites of EPA and DHA may function as mediators of the vasodilatory and cardioprotective effects of omega-3 fatty acids and could serve as biomarkers in clinical studies investigating the cardiovascular effects of EPA/DHA supplementation. dehydroacetic acid 259-262 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 24797816-1 2014 BACKGROUND: Sorafenib, the drug used as first line treatment for hepatocellular carcinoma (HCC), is metabolized by cytochrome P450 (CYP) 3A4-mediated oxidation and uridine diphosphate glucuronosyl transferase (UGT) 1A9-mediated glucuronidation. Sorafenib 12-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-130 24797816-1 2014 BACKGROUND: Sorafenib, the drug used as first line treatment for hepatocellular carcinoma (HCC), is metabolized by cytochrome P450 (CYP) 3A4-mediated oxidation and uridine diphosphate glucuronosyl transferase (UGT) 1A9-mediated glucuronidation. Sorafenib 12-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 24802986-5 2014 Results demonstrated that scutellarin showed negligible inhibitory effects on the six major CYP isoenzymes in human/rat liver microsomes with almost all of the IC50 values exceeding 100 muM, whereas it showed values of 63.8 muM for CYP2C19 in human liver microsomes, and 63.1 and 85.6 muM for CYP2C7 and CYP2C79 in rat liver microsomes, respectively. scutellarin 26-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 24790817-15 2014 Amiodarone was the CYP inhibitor most frequently coprescribed. Amiodarone 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 24304147-0 2014 Metabolism and clinical pharmacokinetics of 2-methyl-n-(2"-(pyrrolidinyl-1-ylsulfonyl)-n-[1,1"-biphenyl]-4-yl)propran-1-amine: insights into monoamine oxidase- and CYP-mediated disposition by integration of in vitro ADME tools. 2-methyl-n-(2"-(pyrrolidinyl-1-ylsulfonyl)-n-[1,1"-biphenyl]-4-yl)propran-1-amine 44-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-167 24304147-2 2014 In early discovery stages, 2-methyl-N-(2"-(pyrrolidinyl-1-ylsulfonyl)-[1,1"-biphenyl]-4-yl)propan-1-amine (PBPA) demonstrated monoamine oxidase A (MAO-A) and cytochrome P450 (CYP)-mediated clearance. 2-methyl-N-((2'-(pyrrolidin-1-ylsulfonyl)biphenyl-4-yl)methyl)propan-1-amine 27-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-173 24304147-2 2014 In early discovery stages, 2-methyl-N-(2"-(pyrrolidinyl-1-ylsulfonyl)-[1,1"-biphenyl]-4-yl)propan-1-amine (PBPA) demonstrated monoamine oxidase A (MAO-A) and cytochrome P450 (CYP)-mediated clearance. 2-methyl-N-((2'-(pyrrolidin-1-ylsulfonyl)biphenyl-4-yl)methyl)propan-1-amine 27-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-178 24304147-2 2014 In early discovery stages, 2-methyl-N-(2"-(pyrrolidinyl-1-ylsulfonyl)-[1,1"-biphenyl]-4-yl)propan-1-amine (PBPA) demonstrated monoamine oxidase A (MAO-A) and cytochrome P450 (CYP)-mediated clearance. 2-methyl-N-((2'-(pyrrolidin-1-ylsulfonyl)biphenyl-4-yl)methyl)propan-1-amine 107-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-173 24304147-2 2014 In early discovery stages, 2-methyl-N-(2"-(pyrrolidinyl-1-ylsulfonyl)-[1,1"-biphenyl]-4-yl)propan-1-amine (PBPA) demonstrated monoamine oxidase A (MAO-A) and cytochrome P450 (CYP)-mediated clearance. 2-methyl-N-((2'-(pyrrolidin-1-ylsulfonyl)biphenyl-4-yl)methyl)propan-1-amine 107-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-178 24760842-0 2014 Overexpression of the steroidogenic enzyme cytochrome P450 side chain cleavage in the ventral tegmental area increases 3alpha,5alpha-THP and reduces long-term operant ethanol self-administration. 5alpha-thp 126-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 24760842-0 2014 Overexpression of the steroidogenic enzyme cytochrome P450 side chain cleavage in the ventral tegmental area increases 3alpha,5alpha-THP and reduces long-term operant ethanol self-administration. Ethanol 167-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 24760842-4 2014 Cytochrome P450 side chain cleavage (P450scc) converts cholesterol to pregnenolone, the rate-limiting enzymatic reaction in neurosteroidogenesis. Cholesterol 55-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24760842-4 2014 Cytochrome P450 side chain cleavage (P450scc) converts cholesterol to pregnenolone, the rate-limiting enzymatic reaction in neurosteroidogenesis. Pregnenolone 70-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24372170-2 2014 However, while in eukaryotes, the cytochrome P450 (CYP)-mediated activation of benzo[a]pyrene (B[a]P) has become a model for metabolism-mediated carcinogenesis, the oxidative degradation of B[a]P by microorganisms is less well studied. Benzo(a)pyrene 79-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 24372170-2 2014 However, while in eukaryotes, the cytochrome P450 (CYP)-mediated activation of benzo[a]pyrene (B[a]P) has become a model for metabolism-mediated carcinogenesis, the oxidative degradation of B[a]P by microorganisms is less well studied. Benzo(a)pyrene 79-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 24567279-1 2014 BACKGROUND: The antiepileptic drug lacosamide has a low potential for drug-drug interactions, but is a substrate and moderate inhibitor of the cytochrome P450 (CYP) enzyme CYP2C19. Lacosamide 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-158 24567279-1 2014 BACKGROUND: The antiepileptic drug lacosamide has a low potential for drug-drug interactions, but is a substrate and moderate inhibitor of the cytochrome P450 (CYP) enzyme CYP2C19. Lacosamide 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-163 24464803-9 2014 Cytochrome P450 (P450) profiling indicated that tofacitinib was mainly metabolized by CYP3A4, with a smaller contribution from CYP2C19. tofacitinib 48-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24165884-0 2014 The effect of single- and multiple-dose etravirine on a drug cocktail of representative cytochrome P450 probes and digoxin in healthy subjects. etravirine 40-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 24165884-1 2014 The effect of etravirine on cytochrome P450 (CYP) enzymes and P-glycoprotein were evaluated in two randomized, crossover trials in healthy subjects. etravirine 14-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 24165884-1 2014 The effect of etravirine on cytochrome P450 (CYP) enzymes and P-glycoprotein were evaluated in two randomized, crossover trials in healthy subjects. etravirine 14-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 24380714-0 2014 In vitro identification of cytochrome P450 isoforms responsible for the metabolism of 1-hydroxyl-2,3,5-trimethoxy-xanthone purified from Halenia elliptica D. Don. 1-hydroxyl-2,3,5-trimethoxy-xanthone 86-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 24642534-4 2014 NADPH-cytochrome P450 oxidoreductase (POR) is a membrane-bound enzyme required for electron transfer to cytochrome P450 (CYP), vital in the processes of the metabolism of drugs and steroid production in humans. Steroids 181-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 24642534-4 2014 NADPH-cytochrome P450 oxidoreductase (POR) is a membrane-bound enzyme required for electron transfer to cytochrome P450 (CYP), vital in the processes of the metabolism of drugs and steroid production in humans. Steroids 181-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 24642534-4 2014 NADPH-cytochrome P450 oxidoreductase (POR) is a membrane-bound enzyme required for electron transfer to cytochrome P450 (CYP), vital in the processes of the metabolism of drugs and steroid production in humans. Steroids 181-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 24342125-2 2014 Inhibition of major human cytochrome P450 enzymes by a standardized hop extract and isolated hop prenylated phenols was evaluated using a fast and efficient assay based on ultrahigh pressure liquid chromatography-tandem mass spectrometry. Phenols 108-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 24618756-2 2014 These flavoproteins function as obligatory intermediates that accept 2 electrons from NAD(P)H with subsequent 1-electron transfers to a variety of cytochrome P450 (CYP) heme proteins within the mitochondria matrix (type I) and the (microsomal) endoplasmic reticulum (type II). nad(p)h 86-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-162 24618756-2 2014 These flavoproteins function as obligatory intermediates that accept 2 electrons from NAD(P)H with subsequent 1-electron transfers to a variety of cytochrome P450 (CYP) heme proteins within the mitochondria matrix (type I) and the (microsomal) endoplasmic reticulum (type II). nad(p)h 86-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-167 24618756-2 2014 These flavoproteins function as obligatory intermediates that accept 2 electrons from NAD(P)H with subsequent 1-electron transfers to a variety of cytochrome P450 (CYP) heme proteins within the mitochondria matrix (type I) and the (microsomal) endoplasmic reticulum (type II). Heme 169-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-162 24618756-2 2014 These flavoproteins function as obligatory intermediates that accept 2 electrons from NAD(P)H with subsequent 1-electron transfers to a variety of cytochrome P450 (CYP) heme proteins within the mitochondria matrix (type I) and the (microsomal) endoplasmic reticulum (type II). Heme 169-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-167 24618756-5 2014 In the microsomal type II system, a diflavin (FAD/FMN)-dependent cytochrome P450 oxidoreductase [NAD(P)H-cytochrome P450 reductase (CPR)] donates electrons to a multitude of heme oxygenases. diflavin 36-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 24618756-5 2014 In the microsomal type II system, a diflavin (FAD/FMN)-dependent cytochrome P450 oxidoreductase [NAD(P)H-cytochrome P450 reductase (CPR)] donates electrons to a multitude of heme oxygenases. Flavin Mononucleotide 50-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 24618756-6 2014 Both flavoenzyme complexes exhibit a commonality of function with all CYP enzymes and are crucial for maintaining a balance of cholesterol and vitamin D metabolites. Cholesterol 127-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 24618756-6 2014 Both flavoenzyme complexes exhibit a commonality of function with all CYP enzymes and are crucial for maintaining a balance of cholesterol and vitamin D metabolites. Vitamin D 143-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 24218006-11 2014 The use of DBS or saliva samples seems feasible for phenotyping of the selected CYP isoforms. dbs 11-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 24464553-2 2014 Although FLX is used as racemate in the clinic, the clinical pharmacokinetics of FLX and its N-demethylation metabolite norfluoxetine (NFLX) show obvious cytochrome P450 (CYP) polymorphism dependency and exhibit marked stereoselectivity. Fluoxetine 81-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 24464553-2 2014 Although FLX is used as racemate in the clinic, the clinical pharmacokinetics of FLX and its N-demethylation metabolite norfluoxetine (NFLX) show obvious cytochrome P450 (CYP) polymorphism dependency and exhibit marked stereoselectivity. Fluoxetine 81-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-174 24464553-2 2014 Although FLX is used as racemate in the clinic, the clinical pharmacokinetics of FLX and its N-demethylation metabolite norfluoxetine (NFLX) show obvious cytochrome P450 (CYP) polymorphism dependency and exhibit marked stereoselectivity. norfluoxetine 120-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 24464553-2 2014 Although FLX is used as racemate in the clinic, the clinical pharmacokinetics of FLX and its N-demethylation metabolite norfluoxetine (NFLX) show obvious cytochrome P450 (CYP) polymorphism dependency and exhibit marked stereoselectivity. norfluoxetine 120-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-174 24464553-2 2014 Although FLX is used as racemate in the clinic, the clinical pharmacokinetics of FLX and its N-demethylation metabolite norfluoxetine (NFLX) show obvious cytochrome P450 (CYP) polymorphism dependency and exhibit marked stereoselectivity. norfluoxetine 135-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 24464553-2 2014 Although FLX is used as racemate in the clinic, the clinical pharmacokinetics of FLX and its N-demethylation metabolite norfluoxetine (NFLX) show obvious cytochrome P450 (CYP) polymorphism dependency and exhibit marked stereoselectivity. norfluoxetine 135-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-174 24387291-2 2014 The importance of CYP enzymes in cardiovascular physiology derives from their ability to metabolize arachidonic acid to epoxyeicosatrienoic and hydroxyeicosatetraenoic acids, which are involved in the maintenance of cardiovascular health, including the regulation of vascular tone, cardiac ion channels and heart contractility. Arachidonic Acid 100-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 24387291-2 2014 The importance of CYP enzymes in cardiovascular physiology derives from their ability to metabolize arachidonic acid to epoxyeicosatrienoic and hydroxyeicosatetraenoic acids, which are involved in the maintenance of cardiovascular health, including the regulation of vascular tone, cardiac ion channels and heart contractility. epoxyeicosatrienoic and hydroxyeicosatetraenoic acids 120-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 23775837-0 2014 Detection of anti-isoniazid and anti-cytochrome P450 antibodies in patients with isoniazid-induced liver failure. Isoniazid 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 24753822-4 2014 Dixon plots of CYP inhibition indicated that thelephoric acid was a competitive inhibitor of CYP1A2 and CYP3A4. thelephoric acid 45-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 24316028-1 2014 Efavirenz (EFV) exhibits interindividual pharmacokinetic variability caused by differences in cytochrome P450 (CYP) expression. efavirenz 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-109 24316028-1 2014 Efavirenz (EFV) exhibits interindividual pharmacokinetic variability caused by differences in cytochrome P450 (CYP) expression. efavirenz 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-114 24753822-0 2014 A Comparison of the In Vitro Inhibitory Effects of Thelephoric Acid and SKF-525A on Human Cytochrome P450 Activity. thelephoric acid 51-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 24753822-0 2014 A Comparison of the In Vitro Inhibitory Effects of Thelephoric Acid and SKF-525A on Human Cytochrome P450 Activity. Proadifen 72-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 24753822-6 2014 Our findings indicate that thelephoric acid may be a novel, non-specific CYP inhibitor, suggesting that it could replace SKF-525A in inhibitory studies designed to investigate the effects of CYP enzymes on the metabolism of given compounds. thelephoric acid 27-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 24753822-6 2014 Our findings indicate that thelephoric acid may be a novel, non-specific CYP inhibitor, suggesting that it could replace SKF-525A in inhibitory studies designed to investigate the effects of CYP enzymes on the metabolism of given compounds. thelephoric acid 27-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-194 24160757-4 2014 CYP-450 enzymes may also contribute to the secondary metabolism of THC, and UDP-glucuronosyltransferases have been identified as capable of catalyzing both primary (CBD, CBN) and secondary (THC, JWH-018, JWH-073) cannabinoid metabolism. Dronabinol 67-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-7 24753822-2 2014 In the present study, the inhibitory effects of polyozellin and thelephoric acid on 9 cytochrome P450 (CYP) family members (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4) were examined in pooled human liver microsomes (HLMs) using a cocktail probe assay. polyozellin 48-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 24753822-2 2014 In the present study, the inhibitory effects of polyozellin and thelephoric acid on 9 cytochrome P450 (CYP) family members (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4) were examined in pooled human liver microsomes (HLMs) using a cocktail probe assay. polyozellin 48-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 24753822-2 2014 In the present study, the inhibitory effects of polyozellin and thelephoric acid on 9 cytochrome P450 (CYP) family members (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4) were examined in pooled human liver microsomes (HLMs) using a cocktail probe assay. thelephoric acid 64-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 24753822-2 2014 In the present study, the inhibitory effects of polyozellin and thelephoric acid on 9 cytochrome P450 (CYP) family members (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4) were examined in pooled human liver microsomes (HLMs) using a cocktail probe assay. thelephoric acid 64-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 24753822-3 2014 Polyozellin exhibited weak inhibitory effects on the activities of all 9 CYPs examined, whereas thelephoric acid exhibited dose- and time-dependent inhibition of all 9 CYP isoforms (IC50 values, 3.2-33.7 muM). polyozellin 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 24040964-0 2014 Influence of inflammation on cardiovascular protective effects of cytochrome P450 epoxygenase-derived epoxyeicosatrienoic acids. epoxyeicosatrienoic acids 102-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 24040964-1 2014 In addition to their role as xenobiotic metabolizing enzymes, cytochrome P450 (CYP) epoxygenases actively contribute to the metabolism of endogenous substances such as arachidonic acid. Arachidonic Acid 168-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 24040964-1 2014 In addition to their role as xenobiotic metabolizing enzymes, cytochrome P450 (CYP) epoxygenases actively contribute to the metabolism of endogenous substances such as arachidonic acid. Arachidonic Acid 168-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 24160757-4 2014 CYP-450 enzymes may also contribute to the secondary metabolism of THC, and UDP-glucuronosyltransferases have been identified as capable of catalyzing both primary (CBD, CBN) and secondary (THC, JWH-018, JWH-073) cannabinoid metabolism. Dronabinol 190-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-7 24160757-4 2014 CYP-450 enzymes may also contribute to the secondary metabolism of THC, and UDP-glucuronosyltransferases have been identified as capable of catalyzing both primary (CBD, CBN) and secondary (THC, JWH-018, JWH-073) cannabinoid metabolism. Cannabinoids 213-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-7 25658126-12 2014 Oxidative bio-activation of this azo dye catalyzed by CYPs and peroxidases leads to generation of proximate genotoxic metabolites (the CYP-catalyzed formation of the benzenediazonium cation and the peroxidase-mediated generation of one-electron oxidation products), which covalently modify DNA both in vitro and in vivo. Azo Compounds 33-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 24168324-0 2014 Cytochrome P450 isoforms are differently up-regulated in aflatoxin B1-exposed human lymphocytes and monocytes. Aflatoxin B1 57-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24634794-3 2014 Here, we report that cytochrome P450 enzymes can serve as efficient catalysts for mediating intramolecular benzylic C-H amination reactions in a variety of arylsulfonyl azide compouds. arylsulfonyl azide 156-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 26161443-2 2014 Enantiomeric separation and quantification of warfarin enantiomers and clinically important major hydroxylation metabolites are essential for drug interaction studies and phenotypic characterization of CYP2C9 and CYP3A4, the major cytochrome P450 (CYP) enzymes involved in warfarin metabolism. Warfarin 46-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 231-246 26161443-2 2014 Enantiomeric separation and quantification of warfarin enantiomers and clinically important major hydroxylation metabolites are essential for drug interaction studies and phenotypic characterization of CYP2C9 and CYP3A4, the major cytochrome P450 (CYP) enzymes involved in warfarin metabolism. Warfarin 46-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-205 24602259-5 2014 However, a detailed literature review of statin metabolism and of SSRI effects on CYP enzymes suggests that escitalopram, citalopram, and paroxetine are almost certain to be safe with all statins, and rosuvastatin, pitavastatin, and pravastatin are almost certain to be safe with all SSRIs. Paroxetine 138-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 24265353-3 2014 Tamoxifen is metabolized by cytochrome P450 (CYP450) enzymes to the more active metabolite endoxifen. Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 24265353-3 2014 Tamoxifen is metabolized by cytochrome P450 (CYP450) enzymes to the more active metabolite endoxifen. Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-51 24265353-3 2014 Tamoxifen is metabolized by cytochrome P450 (CYP450) enzymes to the more active metabolite endoxifen. 4-hydroxy-N-desmethyltamoxifen 91-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 24265353-3 2014 Tamoxifen is metabolized by cytochrome P450 (CYP450) enzymes to the more active metabolite endoxifen. 4-hydroxy-N-desmethyltamoxifen 91-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-51 24265353-4 2014 Several genetic variants in the CYP450 enzymes reduce tamoxifen metabolism, leading to reduced endoxifen levels. Tamoxifen 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-38 24265353-4 2014 Several genetic variants in the CYP450 enzymes reduce tamoxifen metabolism, leading to reduced endoxifen levels. 4-hydroxy-N-desmethyltamoxifen 95-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-38 24265353-8 2014 Genotyping of 33 single nucleotide polymorphisms in CYP450 genes involved in tamoxifen metabolism was carried out using Sequenom MassARRAY. Tamoxifen 77-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-58 25061471-10 2014 Fustin showed moderate inhibitory effects on the CYP2C19 (IC50, 64.3 mug/mL) and weak inhibition of the other CYP isoforms similar to aRVS. fustin 0-6 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 24156555-5 2014 EXPERT OPINION: Rosuvastatin and pitavastatin have favorable pharmacokinetic and safety profiles as their disposition does not depend on or is only marginally influenced by cytochrome P450 (CYP) enzymes, thus potentially reducing the risk of drug-drug interactions of these two statins with other drugs known to inhibit CYP enzymes. Rosuvastatin Calcium 16-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-188 24156555-5 2014 EXPERT OPINION: Rosuvastatin and pitavastatin have favorable pharmacokinetic and safety profiles as their disposition does not depend on or is only marginally influenced by cytochrome P450 (CYP) enzymes, thus potentially reducing the risk of drug-drug interactions of these two statins with other drugs known to inhibit CYP enzymes. Rosuvastatin Calcium 16-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-193 24156555-5 2014 EXPERT OPINION: Rosuvastatin and pitavastatin have favorable pharmacokinetic and safety profiles as their disposition does not depend on or is only marginally influenced by cytochrome P450 (CYP) enzymes, thus potentially reducing the risk of drug-drug interactions of these two statins with other drugs known to inhibit CYP enzymes. Rosuvastatin Calcium 16-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 320-323 24156555-5 2014 EXPERT OPINION: Rosuvastatin and pitavastatin have favorable pharmacokinetic and safety profiles as their disposition does not depend on or is only marginally influenced by cytochrome P450 (CYP) enzymes, thus potentially reducing the risk of drug-drug interactions of these two statins with other drugs known to inhibit CYP enzymes. pitavastatin 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-188 24156555-5 2014 EXPERT OPINION: Rosuvastatin and pitavastatin have favorable pharmacokinetic and safety profiles as their disposition does not depend on or is only marginally influenced by cytochrome P450 (CYP) enzymes, thus potentially reducing the risk of drug-drug interactions of these two statins with other drugs known to inhibit CYP enzymes. pitavastatin 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-193 24156555-5 2014 EXPERT OPINION: Rosuvastatin and pitavastatin have favorable pharmacokinetic and safety profiles as their disposition does not depend on or is only marginally influenced by cytochrome P450 (CYP) enzymes, thus potentially reducing the risk of drug-drug interactions of these two statins with other drugs known to inhibit CYP enzymes. pitavastatin 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 320-323 23474437-0 2014 Cytochrome P450-mediated metabolic alterations in preeclampsia evaluated by quantitative steroid signatures. Steroids 89-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24500235-1 2014 Clopidogrel is a prodrug that undergoes extensive enteric clearance and requires two-stage hepatic activation by cytochrome P450 (CYP) enzymes. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 24500235-1 2014 Clopidogrel is a prodrug that undergoes extensive enteric clearance and requires two-stage hepatic activation by cytochrome P450 (CYP) enzymes. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 23474437-2 2014 The association between various cytochrome P450 (CYP)-mediated steroid metabolic markers and preeclampsia risk was therefore investigated. Steroids 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 23474437-2 2014 The association between various cytochrome P450 (CYP)-mediated steroid metabolic markers and preeclampsia risk was therefore investigated. Steroids 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 23474437-3 2014 The serum levels of 54 CYP-mediated regioselective hydroxysteroids and their substrates were quantitatively evaluated from both pregnant women with preeclampsia (n=30; age, 30.8+-4.5 years) and normotensive controls (n=30; age, 31.0+-3.5 years), who were similar with respect to maternal age, gestational age, and body mass index. Hydroxysteroids 51-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 23474437-8 2014 Our metabolic profiling suggests the CYP-mediated alterations in steroid metabolism and hydroxylation in pregnancy-induced hypertension. Steroids 65-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 25343299-3 2014 The aim of this study was to investigate the inhibitory effects of cedrol, beta-cedrene, and thujopsene on the activities of eight major human cytochrome P-450 (CYP) enzymes using human liver microsomes to assess potential beta-cedrene-, cedrol-, and thujopsene-drug interactions. cedrene 75-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 25343299-3 2014 The aim of this study was to investigate the inhibitory effects of cedrol, beta-cedrene, and thujopsene on the activities of eight major human cytochrome P-450 (CYP) enzymes using human liver microsomes to assess potential beta-cedrene-, cedrol-, and thujopsene-drug interactions. thujopsene 93-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 25343291-11 2014 These results indicate that S-sibutramine was more rapidly metabolized by CYP isoforms than R-sibutramine, and that enantioselective metabolism needs to be considered in drug interactions involving sibutramine and co-administered drugs. sibutramine 28-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 25343291-11 2014 These results indicate that S-sibutramine was more rapidly metabolized by CYP isoforms than R-sibutramine, and that enantioselective metabolism needs to be considered in drug interactions involving sibutramine and co-administered drugs. sibutramine 30-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 25343291-0 2014 Enantioselective N-demethylation and hydroxylation of sibutramine in human liver microsomes and recombinant cytochrome p-450 isoforms. Nitrogen 17-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 25343291-0 2014 Enantioselective N-demethylation and hydroxylation of sibutramine in human liver microsomes and recombinant cytochrome p-450 isoforms. sibutramine 54-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 25343299-3 2014 The aim of this study was to investigate the inhibitory effects of cedrol, beta-cedrene, and thujopsene on the activities of eight major human cytochrome P-450 (CYP) enzymes using human liver microsomes to assess potential beta-cedrene-, cedrol-, and thujopsene-drug interactions. thujopsene 93-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 25343291-1 2014 The enantioselective metabolism of sibutramine was examined using human liver microsomes (HLM) and recombinant cytochrome P-450 (CYP) isoforms. sibutramine 35-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 25343291-1 2014 The enantioselective metabolism of sibutramine was examined using human liver microsomes (HLM) and recombinant cytochrome P-450 (CYP) isoforms. sibutramine 35-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-132 25343299-3 2014 The aim of this study was to investigate the inhibitory effects of cedrol, beta-cedrene, and thujopsene on the activities of eight major human cytochrome P-450 (CYP) enzymes using human liver microsomes to assess potential beta-cedrene-, cedrol-, and thujopsene-drug interactions. cedrol 67-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 25343299-3 2014 The aim of this study was to investigate the inhibitory effects of cedrol, beta-cedrene, and thujopsene on the activities of eight major human cytochrome P-450 (CYP) enzymes using human liver microsomes to assess potential beta-cedrene-, cedrol-, and thujopsene-drug interactions. cedrol 238-244 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 25343299-3 2014 The aim of this study was to investigate the inhibitory effects of cedrol, beta-cedrene, and thujopsene on the activities of eight major human cytochrome P-450 (CYP) enzymes using human liver microsomes to assess potential beta-cedrene-, cedrol-, and thujopsene-drug interactions. cedrol 67-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 25343299-3 2014 The aim of this study was to investigate the inhibitory effects of cedrol, beta-cedrene, and thujopsene on the activities of eight major human cytochrome P-450 (CYP) enzymes using human liver microsomes to assess potential beta-cedrene-, cedrol-, and thujopsene-drug interactions. cedrene 75-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 25343299-3 2014 The aim of this study was to investigate the inhibitory effects of cedrol, beta-cedrene, and thujopsene on the activities of eight major human cytochrome P-450 (CYP) enzymes using human liver microsomes to assess potential beta-cedrene-, cedrol-, and thujopsene-drug interactions. thujopsene 251-261 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 24233717-0 2013 Iron(IV)hydroxide pK(a) and the role of thiolate ligation in C-H bond activation by cytochrome P450. thiolate 40-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 24184833-6 2013 Chemical inhibitors of cytochrome P450 (CYP450) and individual human recombinant CYP450 enzyme were used to identify the CYP450 isozymes involved in the formation of each metabolite of alisol A. alisol A 185-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 24184833-6 2013 Chemical inhibitors of cytochrome P450 (CYP450) and individual human recombinant CYP450 enzyme were used to identify the CYP450 isozymes involved in the formation of each metabolite of alisol A. alisol A 185-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-46 24184833-6 2013 Chemical inhibitors of cytochrome P450 (CYP450) and individual human recombinant CYP450 enzyme were used to identify the CYP450 isozymes involved in the formation of each metabolite of alisol A. alisol A 185-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-87 24184833-6 2013 Chemical inhibitors of cytochrome P450 (CYP450) and individual human recombinant CYP450 enzyme were used to identify the CYP450 isozymes involved in the formation of each metabolite of alisol A. alisol A 185-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-87 24060875-1 2013 Rifampin is a potent inducer of cytochrome P450 (CYP) enzymes and transporters. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 24060875-1 2013 Rifampin is a potent inducer of cytochrome P450 (CYP) enzymes and transporters. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 23974699-0 2013 Chronological effects of rifampicin discontinuation on cytochrome P450 activity in healthy Japanese volunteers, using the cocktail method. Rifampin 25-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 23770199-5 2013 In primary human hepatocytes, clopidogrel (10 and 100 muM) was cytotoxic only after cytochrome P450 (CYP) induction by rifampicin. Clopidogrel 30-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 23770199-5 2013 In primary human hepatocytes, clopidogrel (10 and 100 muM) was cytotoxic only after cytochrome P450 (CYP) induction by rifampicin. Clopidogrel 30-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 23770199-5 2013 In primary human hepatocytes, clopidogrel (10 and 100 muM) was cytotoxic only after cytochrome P450 (CYP) induction by rifampicin. Rifampin 119-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 23770199-5 2013 In primary human hepatocytes, clopidogrel (10 and 100 muM) was cytotoxic only after cytochrome P450 (CYP) induction by rifampicin. Rifampin 119-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 24378836-3 2013 There are growing evidences that clopidogrel response variability is associated with cytochrome P450 (CYP) enzyme genetic polymorphisms, primarily CYP2C19 which is responsible for the conversion of clopidogrel into its active metabolite. Clopidogrel 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 24378836-3 2013 There are growing evidences that clopidogrel response variability is associated with cytochrome P450 (CYP) enzyme genetic polymorphisms, primarily CYP2C19 which is responsible for the conversion of clopidogrel into its active metabolite. Clopidogrel 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 24378836-3 2013 There are growing evidences that clopidogrel response variability is associated with cytochrome P450 (CYP) enzyme genetic polymorphisms, primarily CYP2C19 which is responsible for the conversion of clopidogrel into its active metabolite. Clopidogrel 198-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 24378836-3 2013 There are growing evidences that clopidogrel response variability is associated with cytochrome P450 (CYP) enzyme genetic polymorphisms, primarily CYP2C19 which is responsible for the conversion of clopidogrel into its active metabolite. Clopidogrel 198-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 24378837-2 2013 However, clopidogrel is associated with great variability in antiplatelet response, mostly caused by variable efficacy of cytochrome P450 (CYP) isoforms, which convert clopidogrel to its active metabolite in a two-step process. Clopidogrel 9-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 24378837-2 2013 However, clopidogrel is associated with great variability in antiplatelet response, mostly caused by variable efficacy of cytochrome P450 (CYP) isoforms, which convert clopidogrel to its active metabolite in a two-step process. Clopidogrel 9-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 24378837-2 2013 However, clopidogrel is associated with great variability in antiplatelet response, mostly caused by variable efficacy of cytochrome P450 (CYP) isoforms, which convert clopidogrel to its active metabolite in a two-step process. Clopidogrel 168-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 24378837-2 2013 However, clopidogrel is associated with great variability in antiplatelet response, mostly caused by variable efficacy of cytochrome P450 (CYP) isoforms, which convert clopidogrel to its active metabolite in a two-step process. Clopidogrel 168-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 25278738-5 2013 Genes encoding the liver cytochrome P-450 (CYP) enzymes that are involved with the metabolism of methadone (CYP2B6, 3A4 and 2C19) were selected and genotyped in this cohort. Methadone 97-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 25278738-5 2013 Genes encoding the liver cytochrome P-450 (CYP) enzymes that are involved with the metabolism of methadone (CYP2B6, 3A4 and 2C19) were selected and genotyped in this cohort. Methadone 97-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 23988488-0 2013 Studies on the in vivo contribution of human cytochrome P450s to the hepatic metabolism of glaucine, a new drug of abuse. glaucine 91-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 24523110-4 2014 The great diversity of reactions catalyzed by CYP enzymes appears to be based on two unique properties of these heme proteins, the ability of their iron to exist under multiple oxidation states with different reactivities and a flexible active site that can accommodate a wide variety of substrates. Heme 112-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 24523110-4 2014 The great diversity of reactions catalyzed by CYP enzymes appears to be based on two unique properties of these heme proteins, the ability of their iron to exist under multiple oxidation states with different reactivities and a flexible active site that can accommodate a wide variety of substrates. Iron 148-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 24312617-4 2013 We investigated four CYP subfamilies (CYP1A, CYP2D, CYP2C, and CYP3A) that are involved in 90% of all metabolic drug transformations and identified four amino acid interaction networks associated with specific CYP functionalities, i.e., membrane binding, heme binding, catalytic activity, and dimerization. Heme 255-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 24312617-4 2013 We investigated four CYP subfamilies (CYP1A, CYP2D, CYP2C, and CYP3A) that are involved in 90% of all metabolic drug transformations and identified four amino acid interaction networks associated with specific CYP functionalities, i.e., membrane binding, heme binding, catalytic activity, and dimerization. Heme 255-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 24233717-1 2013 Cytochrome P450 enzymes activate oxygen at heme iron centers to oxidize relatively inert substrate carbon-hydrogen bonds. Oxygen 33-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24233717-1 2013 Cytochrome P450 enzymes activate oxygen at heme iron centers to oxidize relatively inert substrate carbon-hydrogen bonds. Heme 43-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24233717-1 2013 Cytochrome P450 enzymes activate oxygen at heme iron centers to oxidize relatively inert substrate carbon-hydrogen bonds. Iron 48-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24233717-1 2013 Cytochrome P450 enzymes activate oxygen at heme iron centers to oxidize relatively inert substrate carbon-hydrogen bonds. Carbon 99-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24233717-1 2013 Cytochrome P450 enzymes activate oxygen at heme iron centers to oxidize relatively inert substrate carbon-hydrogen bonds. Hydrogen 106-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24117377-0 2013 Cytochrome P450 modified polycrystalline indium tin oxide film as a drug metabolizing electrochemical biosensor with a simple configuration. polycrystalline indium tin oxide 25-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24207099-1 2013 INTRODUCTION: The cytochrome P450 (CYP) enzymes are a class of heme-containing enzymes involved in phase I metabolism of a large number of xenobiotics. Heme 63-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 24207099-1 2013 INTRODUCTION: The cytochrome P450 (CYP) enzymes are a class of heme-containing enzymes involved in phase I metabolism of a large number of xenobiotics. Heme 63-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 24117377-2 2013 We have successfully detected the direct electron transfer (DET) from a human CYP layer or a CYP microsome adsorbed on a bare indium tin oxide (ITO) film electrode without any modification layers and applied it to drug metabolism evaluation. indium tin oxide 126-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 24117377-2 2013 We have successfully detected the direct electron transfer (DET) from a human CYP layer or a CYP microsome adsorbed on a bare indium tin oxide (ITO) film electrode without any modification layers and applied it to drug metabolism evaluation. indium tin oxide 144-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 24117377-4 2013 CYP on polycrystalline ITO film enhanced the electron transfer rate of oxygen reduction about fifteen times more than with amorphous film. polycrystalline ito 7-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 24117377-4 2013 CYP on polycrystalline ITO film enhanced the electron transfer rate of oxygen reduction about fifteen times more than with amorphous film. Oxygen 71-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 24117377-5 2013 The polycrystalline ITO film was a suitable electrode for the adsorption of CYP proteins while maintaining efficient DET and enzymatic activity, probably because of its larger surface area and negatively charged surface. polycrystalline ito 4-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 24117377-6 2013 The oxygen reduction current at the polycrystalline ITO film electrodes had increased 3- to 4-fold, specifically coupled with the oxidation of drugs (testosterone and quinidine) by the monooxygenase activity of CYP. Oxygen 4-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-214 24117377-6 2013 The oxygen reduction current at the polycrystalline ITO film electrodes had increased 3- to 4-fold, specifically coupled with the oxidation of drugs (testosterone and quinidine) by the monooxygenase activity of CYP. polycrystalline 36-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-214 24117377-6 2013 The oxygen reduction current at the polycrystalline ITO film electrodes had increased 3- to 4-fold, specifically coupled with the oxidation of drugs (testosterone and quinidine) by the monooxygenase activity of CYP. Testosterone 150-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-214 24117377-6 2013 The oxygen reduction current at the polycrystalline ITO film electrodes had increased 3- to 4-fold, specifically coupled with the oxidation of drugs (testosterone and quinidine) by the monooxygenase activity of CYP. Quinidine 167-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-214 24117377-7 2013 In contrast, the oxygen reduction current completely disappeared in the presence of the CYP inhibitor (ketoconazole). Oxygen 17-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 24117377-7 2013 In contrast, the oxygen reduction current completely disappeared in the presence of the CYP inhibitor (ketoconazole). Ketoconazole 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 24117377-9 2013 These results indicate that the CYP modified polycrystalline ITO electrode offers the potential for electrochemically evaluating CYP activity for drug metabolism with a simple configuration. polycrystalline 45-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 24117377-9 2013 These results indicate that the CYP modified polycrystalline ITO electrode offers the potential for electrochemically evaluating CYP activity for drug metabolism with a simple configuration. polycrystalline 45-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-132 24113184-3 2013 In the present study, we demonstrate that gp120 and methamphetamine (MA) causes apoptotic cell death by inducing oxidative stress through the cytochrome P450 (CYP) and NADPH oxidase (NOX) pathways. Methamphetamine 52-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 24096258-0 2013 Acute effects of red wine on cytochrome P450 eicosanoids and blood pressure in men. Eicosanoids 45-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). Eicosanoids 25-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). Eicosanoids 25-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). Arachidonic Acid 50-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). Arachidonic Acid 50-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). epoxyeicosatrienoic acids 87-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). epoxyeicosatrienoic acids 87-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). eets 114-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). eets 114-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). 20-hydroxyeicosatrieneoic acid 144-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). 20-hydroxyeicosatrieneoic acid 144-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). 20-Hete 175-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24096258-2 2013 Cytochrome P450 (CYP450) eicosanoids derived from arachidonic acid include vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-hydroxyeicosatrienoic acid (20-HETE). 20-Hete 175-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 24113184-11 2013 Furthermore, we showed that DAS, DPI, and DFO completely abolished apoptosis and cell death, suggesting the involvement of CYP and NOX pathways in ROS-mediated apoptotic cell death. Deferoxamine 42-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 24113184-11 2013 Furthermore, we showed that DAS, DPI, and DFO completely abolished apoptosis and cell death, suggesting the involvement of CYP and NOX pathways in ROS-mediated apoptotic cell death. Reactive Oxygen Species 147-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 24113184-3 2013 In the present study, we demonstrate that gp120 and methamphetamine (MA) causes apoptotic cell death by inducing oxidative stress through the cytochrome P450 (CYP) and NADPH oxidase (NOX) pathways. Methamphetamine 52-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-162 23987522-2 2013 Cytochrome-P450-mediated metabolism of each leads to the generation of pharmacologically active (omega)- and (omega-1)-monohydroxyl metabolites that retain high affinity for cannabinoid type-1 receptors, exhibit Delta(9)-THC-like effects in rodents, and are conjugated with glucuronic acid prior to excretion in human urine. (omega-1)-monohydroxyl 109-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23987522-2 2013 Cytochrome-P450-mediated metabolism of each leads to the generation of pharmacologically active (omega)- and (omega-1)-monohydroxyl metabolites that retain high affinity for cannabinoid type-1 receptors, exhibit Delta(9)-THC-like effects in rodents, and are conjugated with glucuronic acid prior to excretion in human urine. Dronabinol 212-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23987522-2 2013 Cytochrome-P450-mediated metabolism of each leads to the generation of pharmacologically active (omega)- and (omega-1)-monohydroxyl metabolites that retain high affinity for cannabinoid type-1 receptors, exhibit Delta(9)-THC-like effects in rodents, and are conjugated with glucuronic acid prior to excretion in human urine. Glucuronic Acid 274-289 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23770984-2 2013 This study was designed to explore the potential effects of pungent ginger components, 6-, 8-, and 10-gingerol, on human cytochrome P450 (CYP450) enzymes that are responsible for the metabolism of many prescription drugs. 6-, 8-, and 10-gingerol 87-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-136 25505563-0 2013 Enantioselectivity in the cytochrome P450-dependent conversion of tegafur to 5-fluorouracil in human liver microsomes. Tegafur 66-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 25505563-0 2013 Enantioselectivity in the cytochrome P450-dependent conversion of tegafur to 5-fluorouracil in human liver microsomes. Fluorouracil 77-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Tegafur 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Tegafur 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-159 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Tegafur 9-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Tegafur 9-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-159 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Fluorouracil 29-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Fluorouracil 29-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-159 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Fluorouracil 45-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Fluorouracil 45-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-159 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Fluorouracil 111-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 25505563-1 2013 Tegafur (FT) is a prodrug of 5-fluorouracil (5-FU) used in cancer chemotherapy, and the bioactivation of FT to 5-FU is mainly catalyzed by cytochrome P450 (CYP) in hepatic microsomes. Fluorouracil 111-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-159 23852652-0 2013 Effects of cytochrome P450 inhibitors and inducers on the metabolism and pharmacokinetics of ospemifene. Ospemifene 93-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-26 23852652-1 2013 PURPOSE: The objectives were to determine the cytochrome P450 (CYP) enzymes involved in the metabolism of ospemifene and its main hydroxylated metabolites and to examine the effects of CYP inhibitors and inducers on ospemifene pharmacokinetics. Ospemifene 106-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 23852652-1 2013 PURPOSE: The objectives were to determine the cytochrome P450 (CYP) enzymes involved in the metabolism of ospemifene and its main hydroxylated metabolites and to examine the effects of CYP inhibitors and inducers on ospemifene pharmacokinetics. Ospemifene 106-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 23852652-1 2013 PURPOSE: The objectives were to determine the cytochrome P450 (CYP) enzymes involved in the metabolism of ospemifene and its main hydroxylated metabolites and to examine the effects of CYP inhibitors and inducers on ospemifene pharmacokinetics. Ospemifene 216-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 23852652-4 2013 RESULTS: Although several CYP inhibitors decreased the in vitro formation of ospemifene metabolites, none of them completely blocked metabolism. Ospemifene 77-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 23852652-7 2013 CONCLUSIONS: The clinical pharmacokinetic findings and in vitro data suggest that CYP3A4 is important for ospemifene metabolism, but other CYP isoforms and metabolic pathways also contribute. Ospemifene 106-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 23770984-2 2013 This study was designed to explore the potential effects of pungent ginger components, 6-, 8-, and 10-gingerol, on human cytochrome P450 (CYP450) enzymes that are responsible for the metabolism of many prescription drugs. 6-, 8-, and 10-gingerol 87-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-144 23770984-9 2013 CONCLUSION: 6-, 8-, and 10-gingerol suppress human cytochrome P450 activity, while 8- and 10-gingerol inhibit CYP3A4 expression. 6-, 8-, and 10-gingerol 12-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-66 23903277-6 2013 Lipophilic statins, such as atorvastatin, are predominantly metabolized by CYP3A4 and may interfere with CYP activation of clopidogrel, contrary to what happens with hydrophilic statins, such as rosuvastin or pravastatin. Clopidogrel 123-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 23877932-0 2013 Effects of beta-lapachone, a new anticancer candidate, on cytochrome P450-mediated drug metabolism. beta-lapachone 11-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 23877932-1 2013 PURPOSE: This study aimed to assess the potential inhibitory effects of beta-lapachone, a new anticancer candidate, on the activities of the cytochrome P450 (CYP450) enzymes in vitro. beta-lapachone 72-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-156 23877932-1 2013 PURPOSE: This study aimed to assess the potential inhibitory effects of beta-lapachone, a new anticancer candidate, on the activities of the cytochrome P450 (CYP450) enzymes in vitro. beta-lapachone 72-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-164 23877932-6 2013 CONCLUSIONS: These results suggest that pharmacological drug-drug interactions might occur between beta-lapachone and drugs co-administered with it, which are extensively metabolized by CYP450 enzymes, and thus, careful observation is required in clinical pharmacokinetic studies. beta-lapachone 99-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-192 23835282-0 2013 Selective inhibition of the cytochrome P450 isoform by hyperoside and its potent inhibition of CYP2D6. hyperoside 55-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 23835282-2 2013 In the present study, we investigated potential herb-drug inhibitory effects of hyperoside on nine cytochrome P450 (CYP) isoforms in pooled human liver microsomes (HLMs) and human recombinant cDNA expressed CYP using a cocktail probe assay. hyperoside 80-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-114 23835282-2 2013 In the present study, we investigated potential herb-drug inhibitory effects of hyperoside on nine cytochrome P450 (CYP) isoforms in pooled human liver microsomes (HLMs) and human recombinant cDNA expressed CYP using a cocktail probe assay. hyperoside 80-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 23903277-2 2013 Clopidogrel is a pro-drug which requires hepatic cytochrome P450 (CYP) metabolic activation to produce the active metabolite that inhibits platelet aggregation. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 23903277-2 2013 Clopidogrel is a pro-drug which requires hepatic cytochrome P450 (CYP) metabolic activation to produce the active metabolite that inhibits platelet aggregation. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 23903277-5 2013 Also drug-drug interactions that influence the function of CYP isoenzymes may affect the response to clopidogrel. Clopidogrel 101-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 23903277-6 2013 Lipophilic statins, such as atorvastatin, are predominantly metabolized by CYP3A4 and may interfere with CYP activation of clopidogrel, contrary to what happens with hydrophilic statins, such as rosuvastin or pravastatin. Atorvastatin 28-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 23903277-6 2013 Lipophilic statins, such as atorvastatin, are predominantly metabolized by CYP3A4 and may interfere with CYP activation of clopidogrel, contrary to what happens with hydrophilic statins, such as rosuvastin or pravastatin. Pravastatin 209-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 27536445-2 2013 This study investigated the human metabolites of 4-AP and the cytochrome P450 (CYP450) pathways responsible for 4-AP metabolism. 4-Aminopyridine 112-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 25580095-0 2013 DESIGN, SYNTHESIS, AND EVALUATION OF CARBAZOLE ANALOGS AS POTENTIAL CYTOCHROME P450 INHIBITORS. carbazole 37-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 23850985-0 2013 Interactions of sesquiterpenes zederone and germacrone with the human cytochrome P450 system. Sesquiterpenes 16-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 23850985-0 2013 Interactions of sesquiterpenes zederone and germacrone with the human cytochrome P450 system. zederone 31-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 23850985-0 2013 Interactions of sesquiterpenes zederone and germacrone with the human cytochrome P450 system. germacrone 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 23850985-1 2013 Misclassification of Curcuma species (family Zingiberaceae) may lead to unwanted human exposure to Curcuma elata sesquiterpenes zederone and germacrone which have caused hepatotoxicity and changes in CYP expression in laboratory animals. Sesquiterpenes 113-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-203 23850985-1 2013 Misclassification of Curcuma species (family Zingiberaceae) may lead to unwanted human exposure to Curcuma elata sesquiterpenes zederone and germacrone which have caused hepatotoxicity and changes in CYP expression in laboratory animals. germacrone 141-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-203 23850985-6 2013 CYP inhibition studies with pooled human liver microsomes (HLMs) indicated that zederone and germacrone moderately inhibited CYP2B6 and CYP3A4 activities in vitro, with IC50 values below 10 muM. zederone 80-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 23850985-6 2013 CYP inhibition studies with pooled human liver microsomes (HLMs) indicated that zederone and germacrone moderately inhibited CYP2B6 and CYP3A4 activities in vitro, with IC50 values below 10 muM. germacrone 93-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 23561191-0 2013 Hepatic biotransformation of alkylresorcinols is mediated via cytochrome P450 and beta-oxidation: a proof of concept study. alkylresorcinols 29-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 27536445-2 2013 This study investigated the human metabolites of 4-AP and the cytochrome P450 (CYP450) pathways responsible for 4-AP metabolism. 4-Aminopyridine 112-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-85 23554079-0 2013 Effect of bixin and norbixin on the expression of cytochrome P450 in HepG2 cell line. bixin 10-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 23554079-0 2013 Effect of bixin and norbixin on the expression of cytochrome P450 in HepG2 cell line. norbixin 20-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 23554079-3 2013 In this study, the ability of bixin and norbixin to induce the cytochrome P450 (CYP) enzymes was assessed in HepG2 human hepatoma cell line. bixin 30-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 23554079-3 2013 In this study, the ability of bixin and norbixin to induce the cytochrome P450 (CYP) enzymes was assessed in HepG2 human hepatoma cell line. bixin 30-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 23554079-3 2013 In this study, the ability of bixin and norbixin to induce the cytochrome P450 (CYP) enzymes was assessed in HepG2 human hepatoma cell line. norbixin 40-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 23554079-3 2013 In this study, the ability of bixin and norbixin to induce the cytochrome P450 (CYP) enzymes was assessed in HepG2 human hepatoma cell line. norbixin 40-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 23879227-1 2013 Telaprevir (TVR) effects on P-glycloprotein and cytochrome P450 (CYP) may significantly elevate serum levels of morphine and methadone. telaprevir 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 23879227-1 2013 Telaprevir (TVR) effects on P-glycloprotein and cytochrome P450 (CYP) may significantly elevate serum levels of morphine and methadone. telaprevir 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 23879227-1 2013 Telaprevir (TVR) effects on P-glycloprotein and cytochrome P450 (CYP) may significantly elevate serum levels of morphine and methadone. telaprevir 12-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 23879227-1 2013 Telaprevir (TVR) effects on P-glycloprotein and cytochrome P450 (CYP) may significantly elevate serum levels of morphine and methadone. telaprevir 12-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 23879227-1 2013 Telaprevir (TVR) effects on P-glycloprotein and cytochrome P450 (CYP) may significantly elevate serum levels of morphine and methadone. Morphine 112-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 23879227-1 2013 Telaprevir (TVR) effects on P-glycloprotein and cytochrome P450 (CYP) may significantly elevate serum levels of morphine and methadone. Morphine 112-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 23879227-1 2013 Telaprevir (TVR) effects on P-glycloprotein and cytochrome P450 (CYP) may significantly elevate serum levels of morphine and methadone. Methadone 125-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 23879227-1 2013 Telaprevir (TVR) effects on P-glycloprotein and cytochrome P450 (CYP) may significantly elevate serum levels of morphine and methadone. Methadone 125-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 23545481-3 2013 Ranks of CLint for CYP-mediated oxidation/dealkylation of MEHP were human liver>rat liver>human intestine>rat intestine. mono-(2-ethylhexyl)phthalate 58-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 23545481-7 2013 These species/organ differences in major metabolic pathway and CYP isoforms should be considered for appraisal of the potential adverse health effects of DEHP. Diethylhexyl Phthalate 154-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 24216999-2 2013 Cytochrome P450 (CYP) activity and synthesis of both epoxyeicosatrienoic acids (EETs) and 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE), together with vascular endothelial growth factor (VEGF) and heme oxygenase system (HO) have emerged as important modulators of tumor growth and metastasis. epoxyeicosatrienoic acids 53-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23851904-0 2013 GC-MS-based quantitative signatures of cytochrome P450-mediated steroid oxidation induced by rifampicin. Steroids 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 23851904-0 2013 GC-MS-based quantitative signatures of cytochrome P450-mediated steroid oxidation induced by rifampicin. Rifampin 93-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 23851904-2 2013 METHODS: Urinary steroid profiling based on gas chromatography-mass spectrometry (GC-MS) was used for simultaneous quantification of CYP-mediated regioselective hydroxysteroids and their substrates, including 26 androgens, 9 estrogens, 5 progestins, and 7 corticoids. Steroids 17-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 23851904-3 2013 The quantitative data were visualized using a hierarchically clustered heat map to allow identification of CYP-mediated steroid signatures. Steroids 120-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-110 23851904-4 2013 Twelve healthy subjects were orally administered 600 mg of rifampicin a day for 7 days, and their CYP enzyme activity was evaluated. Rifampin 59-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 23851904-8 2013 CONCLUSIONS: This CYP-mediated steroid signature profile allows simultaneous assessment of CYP1A, CYP1B, CYP2C, CYP3A, CYP11B, CYP17A, CYP19A, and CYP21A in urine samples. Steroids 31-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 23880855-0 2013 Effects of ospemifene on drug metabolism mediated by cytochrome P450 enzymes in humans in vitro and in vivo. Ospemifene 11-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 23880855-1 2013 The objective of these investigations was to determine the possible effects of the novel selective estrogen receptor modulator, ospemifene, on cytochrome P450 (CYP)-mediated drug metabolism. Ospemifene 128-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-158 23880855-1 2013 The objective of these investigations was to determine the possible effects of the novel selective estrogen receptor modulator, ospemifene, on cytochrome P450 (CYP)-mediated drug metabolism. Ospemifene 128-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-163 23880855-2 2013 Ospemifene underwent testing for possible effects on CYP enzyme activity in human liver microsomes and in isolated human hepatocytes. Ospemifene 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 23880855-3 2013 Based on the results obtained in vitro, three Phase 1 crossover pharmacokinetic studies were conducted in healthy postmenopausal women to assess the in vivo effects of ospemifene on CYP-mediated drug metabolism. Ospemifene 168-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-185 23880855-8 2013 Therefore, the risk that ospemifene will affect the pharmacokinetics of drugs that are substrates for CYP enzymes is low. Ospemifene 25-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 23479398-3 2013 Efavirenz and protease inhibitor boosted with ritonavir are major components of the antiretroviral therapy and are inducers and/or inhibitors of several cytochrome P450 (CYP) isoforms. Ritonavir 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-168 23479398-3 2013 Efavirenz and protease inhibitor boosted with ritonavir are major components of the antiretroviral therapy and are inducers and/or inhibitors of several cytochrome P450 (CYP) isoforms. Ritonavir 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-173 23983642-5 2013 Cytochrome p450 (CYP) families are implicated in the metabolic activation of BQ- and areca nut-specific nitrosamines. Nitrosamines 104-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23983642-5 2013 Cytochrome p450 (CYP) families are implicated in the metabolic activation of BQ- and areca nut-specific nitrosamines. Nitrosamines 104-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 23643326-2 2013 Since ATRA is metabolized by cytochrome P450 (CYP) enzymes, we sought to identify drug interactions that might be associated with a higher risk for the development of DS in addition to other predictive factors related to the incidence of DS. Tretinoin 6-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 23643326-2 2013 Since ATRA is metabolized by cytochrome P450 (CYP) enzymes, we sought to identify drug interactions that might be associated with a higher risk for the development of DS in addition to other predictive factors related to the incidence of DS. Tretinoin 6-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 24216999-2 2013 Cytochrome P450 (CYP) activity and synthesis of both epoxyeicosatrienoic acids (EETs) and 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE), together with vascular endothelial growth factor (VEGF) and heme oxygenase system (HO) have emerged as important modulators of tumor growth and metastasis. epoxyeicosatrienoic acids 53-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 24216999-2 2013 Cytochrome P450 (CYP) activity and synthesis of both epoxyeicosatrienoic acids (EETs) and 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE), together with vascular endothelial growth factor (VEGF) and heme oxygenase system (HO) have emerged as important modulators of tumor growth and metastasis. eets 80-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24216999-2 2013 Cytochrome P450 (CYP) activity and synthesis of both epoxyeicosatrienoic acids (EETs) and 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE), together with vascular endothelial growth factor (VEGF) and heme oxygenase system (HO) have emerged as important modulators of tumor growth and metastasis. eets 80-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 24216999-2 2013 Cytochrome P450 (CYP) activity and synthesis of both epoxyeicosatrienoic acids (EETs) and 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE), together with vascular endothelial growth factor (VEGF) and heme oxygenase system (HO) have emerged as important modulators of tumor growth and metastasis. -hydroxy-5,8 92-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24216999-2 2013 Cytochrome P450 (CYP) activity and synthesis of both epoxyeicosatrienoic acids (EETs) and 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE), together with vascular endothelial growth factor (VEGF) and heme oxygenase system (HO) have emerged as important modulators of tumor growth and metastasis. -hydroxy-5,8 92-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 24216999-2 2013 Cytochrome P450 (CYP) activity and synthesis of both epoxyeicosatrienoic acids (EETs) and 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE), together with vascular endothelial growth factor (VEGF) and heme oxygenase system (HO) have emerged as important modulators of tumor growth and metastasis. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 134-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 24216999-2 2013 Cytochrome P450 (CYP) activity and synthesis of both epoxyeicosatrienoic acids (EETs) and 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE), together with vascular endothelial growth factor (VEGF) and heme oxygenase system (HO) have emerged as important modulators of tumor growth and metastasis. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 134-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 23632015-1 2013 Cytochrome P450 enzymes have major roles in the metabolism of steroids, drugs, carcinogens, eicosanoids, and numerous other chemicals. Steroids 62-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23632015-1 2013 Cytochrome P450 enzymes have major roles in the metabolism of steroids, drugs, carcinogens, eicosanoids, and numerous other chemicals. Eicosanoids 92-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23632016-1 2013 Cytochrome P450 enzymes primarily catalyze mixed-function oxidation reactions, plus some reductions and rearrangements of oxygenated species, e.g. prostaglandins. Prostaglandins 147-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23619744-8 2013 Surprisingly, the CYP metabolites of AA, epoxyeicosatrienoic acids (EETs), were much less potent activators of TRPV4, and CYP inhibitors did not affect EET production in HCAECs. epoxyeicosatrienoic acids 41-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 23256458-0 2013 The role of human cytochrome P450 enzymes in metabolism of acrylamide in vitro. Acrylamide 59-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 23600686-0 2013 Role of cytochrome P450-mediated arachidonic acid metabolites in the pathogenesis of cardiac hypertrophy. Arachidonic Acid 33-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 23600686-3 2013 The alteration in CYP and sEH expression results in derailed CYP-mediated arachidonic acid (AA) metabolism. Arachidonic Acid 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 23600686-3 2013 The alteration in CYP and sEH expression results in derailed CYP-mediated arachidonic acid (AA) metabolism. Arachidonic Acid 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 26583848-0 2013 The Fe(III)(H2O2) Complex as a Highly Efficient Oxidant in Sulfoxidation Reactions: Revival of an Underrated Oxidant in Cytochrome P450. ferric sulfate 4-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 26583848-0 2013 The Fe(III)(H2O2) Complex as a Highly Efficient Oxidant in Sulfoxidation Reactions: Revival of an Underrated Oxidant in Cytochrome P450. Hydrogen Peroxide 12-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 23149816-2 2013 Clopidogrel is a prodrug which requires a two-step hepatic biotransformation thanks to the cytochrome P450 (CYP450) enzyme system. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 23149816-2 2013 Clopidogrel is a prodrug which requires a two-step hepatic biotransformation thanks to the cytochrome P450 (CYP450) enzyme system. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-114 23149816-3 2013 Genetic polymorphism of CYP450 system (e.g., CYP2C19*2) responsible for altered clopidogrel metabolism is a major cause of high on-treatment platelet reactivity (HTPR), which translates into thrombotic events in stented patients. Clopidogrel 80-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-30 23379642-3 2013 ROS is produced by a number of pathways, including the mitochondrial electron transport chain, cytochrome P450, xanthine oxidase and uncoupled nitric oxide synthase. Reactive Oxygen Species 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 23340899-1 2013 Clotrimazole is an azole fungicide used as a human pharmaceutical that is known to inhibit cytochrome P450 (CYP) enzymatic activities, including several steroidogenic CYP. Clotrimazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 23340899-1 2013 Clotrimazole is an azole fungicide used as a human pharmaceutical that is known to inhibit cytochrome P450 (CYP) enzymatic activities, including several steroidogenic CYP. Clotrimazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 23340899-1 2013 Clotrimazole is an azole fungicide used as a human pharmaceutical that is known to inhibit cytochrome P450 (CYP) enzymatic activities, including several steroidogenic CYP. Clotrimazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-170 23340899-1 2013 Clotrimazole is an azole fungicide used as a human pharmaceutical that is known to inhibit cytochrome P450 (CYP) enzymatic activities, including several steroidogenic CYP. Azoles 7-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 23340899-1 2013 Clotrimazole is an azole fungicide used as a human pharmaceutical that is known to inhibit cytochrome P450 (CYP) enzymatic activities, including several steroidogenic CYP. Azoles 7-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 23340899-1 2013 Clotrimazole is an azole fungicide used as a human pharmaceutical that is known to inhibit cytochrome P450 (CYP) enzymatic activities, including several steroidogenic CYP. Azoles 7-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-170 23619744-8 2013 Surprisingly, the CYP metabolites of AA, epoxyeicosatrienoic acids (EETs), were much less potent activators of TRPV4, and CYP inhibitors did not affect EET production in HCAECs. eets 68-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 23432465-1 2013 Human cytochrome P450 (P450) 2A13 was found to interact with several polycyclic aromatic hydrocarbons (PAHs) to produce Type I binding spectra, including acenaphthene, acenaphthylene, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, and 1-nitropyrene. Polycyclic Aromatic Hydrocarbons 69-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-33 23432465-1 2013 Human cytochrome P450 (P450) 2A13 was found to interact with several polycyclic aromatic hydrocarbons (PAHs) to produce Type I binding spectra, including acenaphthene, acenaphthylene, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, and 1-nitropyrene. Polycyclic Aromatic Hydrocarbons 103-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-33 23432465-1 2013 Human cytochrome P450 (P450) 2A13 was found to interact with several polycyclic aromatic hydrocarbons (PAHs) to produce Type I binding spectra, including acenaphthene, acenaphthylene, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, and 1-nitropyrene. acenaphthene 154-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-33 23432465-1 2013 Human cytochrome P450 (P450) 2A13 was found to interact with several polycyclic aromatic hydrocarbons (PAHs) to produce Type I binding spectra, including acenaphthene, acenaphthylene, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, and 1-nitropyrene. acenaphthylene 168-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-33 23432465-1 2013 Human cytochrome P450 (P450) 2A13 was found to interact with several polycyclic aromatic hydrocarbons (PAHs) to produce Type I binding spectra, including acenaphthene, acenaphthylene, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, and 1-nitropyrene. benzo(c)phenanthrene 184-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-33 23432465-1 2013 Human cytochrome P450 (P450) 2A13 was found to interact with several polycyclic aromatic hydrocarbons (PAHs) to produce Type I binding spectra, including acenaphthene, acenaphthylene, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, and 1-nitropyrene. fluoranthene 206-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-33 23432465-1 2013 Human cytochrome P450 (P450) 2A13 was found to interact with several polycyclic aromatic hydrocarbons (PAHs) to produce Type I binding spectra, including acenaphthene, acenaphthylene, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, and 1-nitropyrene. fluoranthene-2,3-diol 220-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-33 23432465-1 2013 Human cytochrome P450 (P450) 2A13 was found to interact with several polycyclic aromatic hydrocarbons (PAHs) to produce Type I binding spectra, including acenaphthene, acenaphthylene, benzo[c]phenanthrene, fluoranthene, fluoranthene-2,3-diol, and 1-nitropyrene. 1-nitropyrene 247-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-33 22999230-3 2013 Concentrations of doxepin metabolites and isomers, pharmacokinetic modelling and genotyping of the doxepin-metabolizing cytochrome P450 (CYP) enzymes led to the following conclusion: the lethal doxepin concentration of 2100 ng/mL was more likely to have been reached due to drug interactions and genetic peculiarities leading to a reduction of the metabolic capacity and not by an acute (suicidal) overdose. Doxepin 99-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 22999230-3 2013 Concentrations of doxepin metabolites and isomers, pharmacokinetic modelling and genotyping of the doxepin-metabolizing cytochrome P450 (CYP) enzymes led to the following conclusion: the lethal doxepin concentration of 2100 ng/mL was more likely to have been reached due to drug interactions and genetic peculiarities leading to a reduction of the metabolic capacity and not by an acute (suicidal) overdose. Doxepin 99-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 22999230-3 2013 Concentrations of doxepin metabolites and isomers, pharmacokinetic modelling and genotyping of the doxepin-metabolizing cytochrome P450 (CYP) enzymes led to the following conclusion: the lethal doxepin concentration of 2100 ng/mL was more likely to have been reached due to drug interactions and genetic peculiarities leading to a reduction of the metabolic capacity and not by an acute (suicidal) overdose. Doxepin 99-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 22999230-3 2013 Concentrations of doxepin metabolites and isomers, pharmacokinetic modelling and genotyping of the doxepin-metabolizing cytochrome P450 (CYP) enzymes led to the following conclusion: the lethal doxepin concentration of 2100 ng/mL was more likely to have been reached due to drug interactions and genetic peculiarities leading to a reduction of the metabolic capacity and not by an acute (suicidal) overdose. Doxepin 99-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 23299247-2 2013 This study characterized the phase I and phase II metabolites, metabolic kinetics, and cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzymes responsible for the metabolism of jatrorrhizine in human liver microsomes (HLMs). jatrorrhizine 189-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 23466098-0 2013 Cytochrome P450-derived eicosanoids and vascular dysfunction in coronary artery disease patients. Eicosanoids 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23466098-1 2013 OBJECTIVE: Accumulating preclinical and epidemiologic evidence has emerged to suggest that modulation of cytochrome P450 (CYP)-mediated eicosanoid metabolism may be a viable vascular protective therapeutic strategy for the secondary prevention of coronary artery disease (CAD). Eicosanoids 136-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 23466098-1 2013 OBJECTIVE: Accumulating preclinical and epidemiologic evidence has emerged to suggest that modulation of cytochrome P450 (CYP)-mediated eicosanoid metabolism may be a viable vascular protective therapeutic strategy for the secondary prevention of coronary artery disease (CAD). Eicosanoids 136-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 23466098-2 2013 The functional relationship between CYP-derived eicosanoid metabolite levels and vascular dysfunction in humans with established CAD, however, has not been evaluated. Eicosanoids 48-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 23466098-5 2013 Relationships between biomarkers of CYP-mediated eicosanoid metabolism and vascular function phenotypes were evaluated by Pearson"s correlation. Eicosanoids 49-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 23466098-6 2013 RESULTS: A significant inverse association was observed between 20-HETE levels (a biomarker of CYP omega-hydroxylase metabolism) and brachial artery flow-mediated dilation (r = -0.255, p = 0.010). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-98 23514038-3 2013 Brz2001 is a brassinosteroid biosynthesis inhibitor in the less-soluble triazole series of compounds that targets DWARF4, a cytochrome P450 (Cyp450) monooxygenase containing heme and iron. Brz2001 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 23514038-3 2013 Brz2001 is a brassinosteroid biosynthesis inhibitor in the less-soluble triazole series of compounds that targets DWARF4, a cytochrome P450 (Cyp450) monooxygenase containing heme and iron. Brz2001 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-147 23514038-3 2013 Brz2001 is a brassinosteroid biosynthesis inhibitor in the less-soluble triazole series of compounds that targets DWARF4, a cytochrome P450 (Cyp450) monooxygenase containing heme and iron. Brassinosteroids 13-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 23514038-3 2013 Brz2001 is a brassinosteroid biosynthesis inhibitor in the less-soluble triazole series of compounds that targets DWARF4, a cytochrome P450 (Cyp450) monooxygenase containing heme and iron. Brassinosteroids 13-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-147 23514038-3 2013 Brz2001 is a brassinosteroid biosynthesis inhibitor in the less-soluble triazole series of compounds that targets DWARF4, a cytochrome P450 (Cyp450) monooxygenase containing heme and iron. Triazoles 72-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 23299247-2 2013 This study characterized the phase I and phase II metabolites, metabolic kinetics, and cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) enzymes responsible for the metabolism of jatrorrhizine in human liver microsomes (HLMs). jatrorrhizine 189-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-107 23514038-3 2013 Brz2001 is a brassinosteroid biosynthesis inhibitor in the less-soluble triazole series of compounds that targets DWARF4, a cytochrome P450 (Cyp450) monooxygenase containing heme and iron. Triazoles 72-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-147 23514038-3 2013 Brz2001 is a brassinosteroid biosynthesis inhibitor in the less-soluble triazole series of compounds that targets DWARF4, a cytochrome P450 (Cyp450) monooxygenase containing heme and iron. Heme 174-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-147 23319543-0 2013 Estradiol-17beta and its cytochrome P450- and catechol-O-methyltransferase-derived metabolites selectively stimulate production of prostacyclin in uterine artery endothelial cells: role of estrogen receptor-alpha versus estrogen receptor-beta. Epoprostenol 131-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-74 23514038-3 2013 Brz2001 is a brassinosteroid biosynthesis inhibitor in the less-soluble triazole series of compounds that targets DWARF4, a cytochrome P450 (Cyp450) monooxygenase containing heme and iron. Iron 183-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-147 23480885-1 2013 Hepatic cytochrome P450 (CYP) bioactivation of clopidogrel is reduced in subjects with the CYP2C19*2 loss-of-function allele. Clopidogrel 47-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 23480885-1 2013 Hepatic cytochrome P450 (CYP) bioactivation of clopidogrel is reduced in subjects with the CYP2C19*2 loss-of-function allele. Clopidogrel 47-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 23682310-0 2013 How Do Perfluorinated Alkanoic Acids Elicit Cytochrome P450 to Catalyze Methane Hydroxylation? alkanoic acids 22-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 23682310-0 2013 How Do Perfluorinated Alkanoic Acids Elicit Cytochrome P450 to Catalyze Methane Hydroxylation? Methane 72-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 23336137-5 2013 RESULTS: The cytochrome P-450 catalyzed hydroxylation rate decreased as the number of chlorine substitutions increased. Chlorine 86-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 23599745-6 2013 AFB1 is metabolized by cytochrome-P450 enzymes to the reactive intermediate AFB1-8, 9 epoxide (AFBO) which binds to liver cell DNA, resulting in DNA adducts. Aflatoxin B1 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 23599745-6 2013 AFB1 is metabolized by cytochrome-P450 enzymes to the reactive intermediate AFB1-8, 9 epoxide (AFBO) which binds to liver cell DNA, resulting in DNA adducts. afb1-8, 9 epoxide 76-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 23599745-6 2013 AFB1 is metabolized by cytochrome-P450 enzymes to the reactive intermediate AFB1-8, 9 epoxide (AFBO) which binds to liver cell DNA, resulting in DNA adducts. afbo 95-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 23117072-1 2013 BACKGROUND: This study explored the impact of genetic polymorphisms in cytochrome P450 (CYP) enzymes and transporters on the plasma trough concentration of imatinib mesylate (IM) and clinical response in chronic myeloid leukemia (CML). Imatinib Mesylate 156-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 23117072-1 2013 BACKGROUND: This study explored the impact of genetic polymorphisms in cytochrome P450 (CYP) enzymes and transporters on the plasma trough concentration of imatinib mesylate (IM) and clinical response in chronic myeloid leukemia (CML). Imatinib Mesylate 156-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 23406885-5 2013 FINDINGS: We conducted CYP phenotyping in recombinantly expressed systems, and in human liver microsomes, to evaluate CYP isozyme contributions to the metabolism of PiB (carrier) and profiled microsomal and hepatocyte incubations for metabolites. 4-iodobenzenesulfonamide 165-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 22937971-0 2013 Design, synthesis, and evaluation of novel cyclic phosphates of 5-aminosalicylic acid as cytochrome p450-activated prodrugs. cyclic phosphates 43-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 22937971-0 2013 Design, synthesis, and evaluation of novel cyclic phosphates of 5-aminosalicylic acid as cytochrome p450-activated prodrugs. Mesalamine 64-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 22937971-1 2013 Four novel cyclic phosphates of the anti-inflammatory agent 5-aminosalicylic acid (5-ASA) were designed and synthesized as cytochrome P450 (CYP)-activated prodrugs. cyclic phosphates 11-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-138 22937971-1 2013 Four novel cyclic phosphates of the anti-inflammatory agent 5-aminosalicylic acid (5-ASA) were designed and synthesized as cytochrome P450 (CYP)-activated prodrugs. cyclic phosphates 11-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-143 22937971-1 2013 Four novel cyclic phosphates of the anti-inflammatory agent 5-aminosalicylic acid (5-ASA) were designed and synthesized as cytochrome P450 (CYP)-activated prodrugs. Mesalamine 60-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-138 22937971-1 2013 Four novel cyclic phosphates of the anti-inflammatory agent 5-aminosalicylic acid (5-ASA) were designed and synthesized as cytochrome P450 (CYP)-activated prodrugs. Mesalamine 60-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-143 22937971-1 2013 Four novel cyclic phosphates of the anti-inflammatory agent 5-aminosalicylic acid (5-ASA) were designed and synthesized as cytochrome P450 (CYP)-activated prodrugs. Mesalamine 83-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-138 22937971-1 2013 Four novel cyclic phosphates of the anti-inflammatory agent 5-aminosalicylic acid (5-ASA) were designed and synthesized as cytochrome P450 (CYP)-activated prodrugs. Mesalamine 83-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-143 22765278-3 2013 METHODS: Fourteen healthy, male volunteers aged 18 to 45 years were recruited into an open label, two period, single centre study in which CYP enzyme activity was measured by administration of the selectively metabolized probe drugs theophylline (CYP1A2), tolbutamide (CYP2C9), mephenytoin (CYP2C19), debrisoquine (CYP2D6) and dapsone (CYP3A4) on day 1 of the study. Theophylline 233-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 22765278-3 2013 METHODS: Fourteen healthy, male volunteers aged 18 to 45 years were recruited into an open label, two period, single centre study in which CYP enzyme activity was measured by administration of the selectively metabolized probe drugs theophylline (CYP1A2), tolbutamide (CYP2C9), mephenytoin (CYP2C19), debrisoquine (CYP2D6) and dapsone (CYP3A4) on day 1 of the study. Tolbutamide 256-267 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 22765278-3 2013 METHODS: Fourteen healthy, male volunteers aged 18 to 45 years were recruited into an open label, two period, single centre study in which CYP enzyme activity was measured by administration of the selectively metabolized probe drugs theophylline (CYP1A2), tolbutamide (CYP2C9), mephenytoin (CYP2C19), debrisoquine (CYP2D6) and dapsone (CYP3A4) on day 1 of the study. Mephenytoin 278-289 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 22765278-3 2013 METHODS: Fourteen healthy, male volunteers aged 18 to 45 years were recruited into an open label, two period, single centre study in which CYP enzyme activity was measured by administration of the selectively metabolized probe drugs theophylline (CYP1A2), tolbutamide (CYP2C9), mephenytoin (CYP2C19), debrisoquine (CYP2D6) and dapsone (CYP3A4) on day 1 of the study. Debrisoquin 301-313 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 22765278-3 2013 METHODS: Fourteen healthy, male volunteers aged 18 to 45 years were recruited into an open label, two period, single centre study in which CYP enzyme activity was measured by administration of the selectively metabolized probe drugs theophylline (CYP1A2), tolbutamide (CYP2C9), mephenytoin (CYP2C19), debrisoquine (CYP2D6) and dapsone (CYP3A4) on day 1 of the study. Dapsone 327-334 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 23224081-5 2013 Mass spectrometric analysis identified CYP2S1 as a novel macrophage CYP and CYP2S1-containing microsomes generated epoxides of arachidonic, linoleic and eicosapentaenoic acid. Epoxy Compounds 115-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 22809523-3 2013 CYP reactions in vivo require the cofactor NADPH as the source of electrons and an additional enzyme, cytochrome P450 reductase (CPR), as the electron transfer partner; consequently, any laboratory or industrial use of CYPs is limited by the need to supply NADPH and CPR. NADP 43-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 22809523-3 2013 CYP reactions in vivo require the cofactor NADPH as the source of electrons and an additional enzyme, cytochrome P450 reductase (CPR), as the electron transfer partner; consequently, any laboratory or industrial use of CYPs is limited by the need to supply NADPH and CPR. NADP 257-262 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 23215036-1 2013 The explosive Hexahydro-1,3,5-trinitro-1,3,5-triazine (RDX) is known to be degraded aerobically by various isolates of the Rhodococcus species, with denitration being the key step, mediated by Cytochrome P450. cyclonite 14-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-208 23215036-1 2013 The explosive Hexahydro-1,3,5-trinitro-1,3,5-triazine (RDX) is known to be degraded aerobically by various isolates of the Rhodococcus species, with denitration being the key step, mediated by Cytochrome P450. cyclonite 55-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-208 23071008-1 2013 Induction of the cytochrome P450 (CYP) family of enzymes by coadministered compounds can result in drug-drug interactions, as in the case of the coadministration of rifampicin with many CYP3A substrates, including midazolam. Rifampin 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-32 23071008-1 2013 Induction of the cytochrome P450 (CYP) family of enzymes by coadministered compounds can result in drug-drug interactions, as in the case of the coadministration of rifampicin with many CYP3A substrates, including midazolam. Rifampin 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 23071008-1 2013 Induction of the cytochrome P450 (CYP) family of enzymes by coadministered compounds can result in drug-drug interactions, as in the case of the coadministration of rifampicin with many CYP3A substrates, including midazolam. Midazolam 214-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-32 23071008-1 2013 Induction of the cytochrome P450 (CYP) family of enzymes by coadministered compounds can result in drug-drug interactions, as in the case of the coadministration of rifampicin with many CYP3A substrates, including midazolam. Midazolam 214-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 24362992-1 2013 Lanosterol 14a-demethylase CYP51 is the most conserved cytochrome P450 (CYP) and is a part of hepatic cholesterol synthesis. Lanosterol 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 24362992-1 2013 Lanosterol 14a-demethylase CYP51 is the most conserved cytochrome P450 (CYP) and is a part of hepatic cholesterol synthesis. Lanosterol 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 24362992-1 2013 Lanosterol 14a-demethylase CYP51 is the most conserved cytochrome P450 (CYP) and is a part of hepatic cholesterol synthesis. Cholesterol 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 24362992-1 2013 Lanosterol 14a-demethylase CYP51 is the most conserved cytochrome P450 (CYP) and is a part of hepatic cholesterol synthesis. Cholesterol 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 23224081-1 2013 Cytochrome P450 (CYP) epoxygenases metabolize endogenous polyunsaturated fatty acids to their corresponding epoxides, generating bioactive lipid mediators. Fatty Acids, Unsaturated 57-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23224081-1 2013 Cytochrome P450 (CYP) epoxygenases metabolize endogenous polyunsaturated fatty acids to their corresponding epoxides, generating bioactive lipid mediators. Fatty Acids, Unsaturated 57-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 23224081-1 2013 Cytochrome P450 (CYP) epoxygenases metabolize endogenous polyunsaturated fatty acids to their corresponding epoxides, generating bioactive lipid mediators. Epoxy Compounds 108-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23224081-1 2013 Cytochrome P450 (CYP) epoxygenases metabolize endogenous polyunsaturated fatty acids to their corresponding epoxides, generating bioactive lipid mediators. Epoxy Compounds 108-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 23224081-5 2013 Mass spectrometric analysis identified CYP2S1 as a novel macrophage CYP and CYP2S1-containing microsomes generated epoxides of arachidonic, linoleic and eicosapentaenoic acid. arachidonic 127-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 23224081-5 2013 Mass spectrometric analysis identified CYP2S1 as a novel macrophage CYP and CYP2S1-containing microsomes generated epoxides of arachidonic, linoleic and eicosapentaenoic acid. Linoleic Acid 140-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 23224081-5 2013 Mass spectrometric analysis identified CYP2S1 as a novel macrophage CYP and CYP2S1-containing microsomes generated epoxides of arachidonic, linoleic and eicosapentaenoic acid. Eicosapentaenoic Acid 153-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 23587001-1 2013 The mammalian cytochrome P450 (CYP) enzymes play important roles in drug metabolism, steroid biosynthesis, and xenobiotic degradation. Steroids 85-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 23587001-1 2013 The mammalian cytochrome P450 (CYP) enzymes play important roles in drug metabolism, steroid biosynthesis, and xenobiotic degradation. Steroids 85-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 23586999-0 2013 Amphipol trapping of a functional CYP system. amphipol 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 23419353-0 2013 In vitro inhibition and induction of human liver cytochrome P450 enzymes by gentiopicroside: potent effect on CYP2A6. gentiopicroside 76-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 23001793-0 2013 Stereoselective interaction between tetrahydropalmatine enantiomers and CYP enzymes in human liver microsomes. tetrahydropalmatine 36-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 23688132-5 2013 These inhibitors generally target the cytochrome P450 (CYP) enzymes because RA clearance is predominantly mediated by P450s. Tretinoin 76-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 23688132-5 2013 These inhibitors generally target the cytochrome P450 (CYP) enzymes because RA clearance is predominantly mediated by P450s. Tretinoin 76-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 23688133-0 2013 CYP4 enzymes as potential drug targets: focus on enzyme multiplicity, inducers and inhibitors, and therapeutic modulation of 20-hydroxyeicosatetraenoic acid (20-HETE) synthase and fatty acid omega-hydroxylase activities. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 125-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-175 23165864-8 2013 Genetic factors including single nucleotide polymorphisms (SNPs) that change cytochrome P450 (CYP) activity and epigenetic regulation that modifies CYP expression levels may contribute to the changes in pharmacokinetics and adverse drug reactions (ADRs) of thalidomide. Thalidomide 257-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 23165864-8 2013 Genetic factors including single nucleotide polymorphisms (SNPs) that change cytochrome P450 (CYP) activity and epigenetic regulation that modifies CYP expression levels may contribute to the changes in pharmacokinetics and adverse drug reactions (ADRs) of thalidomide. Thalidomide 257-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 23419353-2 2013 However, the inhibitory and inducible effects of GE on the activity of cytochrome P450 (CYP450) are unclear. gentiopicroside 49-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 23419353-2 2013 However, the inhibitory and inducible effects of GE on the activity of cytochrome P450 (CYP450) are unclear. gentiopicroside 49-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-94 23903227-0 2013 Enhancement of quinone hepatotoxicity by cytochrome P450 inhibition. quinone 15-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 23293967-4 2013 We investigated the levels of 8- hydroxydeoxyguanosine (8-OHdG), one of the most sensitive biomarkers for measuring OS and the association between polymorphisms in Cytochrome P450 (CYP) and Glutathione S-Transferase (GST) genes that are known to play a significant role in the activation and detoxification of xenobiotics. 8-ohdg 56-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-184 22426195-2 2013 A potential drug-drug interaction exists between docetaxel and ketoconazole because both agents are metabolized hepatically by the cytochrome P-450 system, and ketoconazole can inhibit p-gp efflux of docetaxel at blood brain barrier. Docetaxel 49-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 22426195-2 2013 A potential drug-drug interaction exists between docetaxel and ketoconazole because both agents are metabolized hepatically by the cytochrome P-450 system, and ketoconazole can inhibit p-gp efflux of docetaxel at blood brain barrier. Ketoconazole 63-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 23483888-5 2013 Our data in centenarians support promotion of cellular detoxification mechanisms through specific modulation of the arachidonic acid metabolic cascade as we underpinned increased concentration of 8,9-EpETrE, suggesting enhanced cytochrome P450 (CYP) enzyme activity. Arachidonic Acid 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 23483888-5 2013 Our data in centenarians support promotion of cellular detoxification mechanisms through specific modulation of the arachidonic acid metabolic cascade as we underpinned increased concentration of 8,9-EpETrE, suggesting enhanced cytochrome P450 (CYP) enzyme activity. Arachidonic Acid 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 245-248 23481074-0 2013 Cytochrome P450 (CYP2C9*2,*3) & vitamin-K epoxide reductase complex (VKORC1 -1639G<A) gene polymorphisms & their effect on acenocoumarol dose in patients with mechanical heart valve replacement. Acenocoumarol 134-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 23481074-1 2013 BACKGROUND & OBJECTIVES: Studies have demonstrated the effect of CYP2C9 (cytochrome P450) and VKORC1 (vitamin K epoxide reductase complex) gene polymorphisms on the dose of acenocoumarol. Adenosine Monophosphate 12-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 23481074-1 2013 BACKGROUND & OBJECTIVES: Studies have demonstrated the effect of CYP2C9 (cytochrome P450) and VKORC1 (vitamin K epoxide reductase complex) gene polymorphisms on the dose of acenocoumarol. Acenocoumarol 177-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 23947696-1 2013 Iron(III)-porphyrin complexes are generally regarded as green catalysts, since they mimic the catalytic center of cytochrome-P450 and widely used as green catalysts for degrading halogenated phenols in wastewater, such as landfill leachates. iron(iii)-porphyrin 0-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 23947696-1 2013 Iron(III)-porphyrin complexes are generally regarded as green catalysts, since they mimic the catalytic center of cytochrome-P450 and widely used as green catalysts for degrading halogenated phenols in wastewater, such as landfill leachates. Phenols 191-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 23903227-4 2013 A drug metabolizing enzyme, cytochrome P450 is closely involved in the hepatotoxicity of therapeutic agents in general, but quinone hepatotoxicity has been considered not to contribute to cytochrome P450 because the structure of quinone is not modified by cytochrome P450 and thus quinone compounds are thought to be metabolized mainly via a conjugation process. quinone 229-236 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 23903227-4 2013 A drug metabolizing enzyme, cytochrome P450 is closely involved in the hepatotoxicity of therapeutic agents in general, but quinone hepatotoxicity has been considered not to contribute to cytochrome P450 because the structure of quinone is not modified by cytochrome P450 and thus quinone compounds are thought to be metabolized mainly via a conjugation process. quinone 229-236 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 23903227-5 2013 However, we have recently shown that quinone hepatotoxicity is enhanced under conditions of cytochrome P450 inhibition, indicating clearly the involvement of cytochrome P450 in quinone hepatotoxicity. quinone 37-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 23903227-5 2013 However, we have recently shown that quinone hepatotoxicity is enhanced under conditions of cytochrome P450 inhibition, indicating clearly the involvement of cytochrome P450 in quinone hepatotoxicity. quinone 37-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-173 23903227-5 2013 However, we have recently shown that quinone hepatotoxicity is enhanced under conditions of cytochrome P450 inhibition, indicating clearly the involvement of cytochrome P450 in quinone hepatotoxicity. quinone 177-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 23903227-5 2013 However, we have recently shown that quinone hepatotoxicity is enhanced under conditions of cytochrome P450 inhibition, indicating clearly the involvement of cytochrome P450 in quinone hepatotoxicity. quinone 177-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-173 23903227-6 2013 Here, we revisit the generally accepted mechanisms of quinone hepatotoxicity and propose the importance of cytochrome P450 systems in quinone-induced hepatotoxicity on the basis of our recent work. quinone 134-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 23186213-1 2012 Cytochrome P450 (P450) 2E1 is the major enzyme that oxidizes N-nitrosodimethylamine [N,N-dimethylnitrosamine (DMN)], a carcinogen and also a representative of some nitrosamines formed endogenously. Dimethylnitrosamine 61-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 23186213-1 2012 Cytochrome P450 (P450) 2E1 is the major enzyme that oxidizes N-nitrosodimethylamine [N,N-dimethylnitrosamine (DMN)], a carcinogen and also a representative of some nitrosamines formed endogenously. Dimethylnitrosamine 85-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 23186213-1 2012 Cytochrome P450 (P450) 2E1 is the major enzyme that oxidizes N-nitrosodimethylamine [N,N-dimethylnitrosamine (DMN)], a carcinogen and also a representative of some nitrosamines formed endogenously. Dimethylnitrosamine 110-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 23186213-1 2012 Cytochrome P450 (P450) 2E1 is the major enzyme that oxidizes N-nitrosodimethylamine [N,N-dimethylnitrosamine (DMN)], a carcinogen and also a representative of some nitrosamines formed endogenously. Nitrosamines 164-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 22834478-6 2012 Whereas positive controls (100 microM omeprazole and 10 microM rifampin) caused the anticipated CYP induction, the highest concentration of mirabegron (10 microM; 37 times plasma C(max)) had minimal effect on CYP1A2 and CYP3A4/5 activity, and CYP1A2 and CYP3A4 mRNA levels in freshly isolated human hepatocytes, suggesting that mirabegron is not an inducer of these enzymes. Rifampin 63-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 23174983-2 2012 The catalytic properties of cytochrome P-450 and soluble methane monooxygenase enzymes are associated with oxo species on mononuclear iron haem and diiron non-haem platforms, respectively. Iron 134-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 23174983-2 2012 The catalytic properties of cytochrome P-450 and soluble methane monooxygenase enzymes are associated with oxo species on mononuclear iron haem and diiron non-haem platforms, respectively. diiron 148-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 23174983-2 2012 The catalytic properties of cytochrome P-450 and soluble methane monooxygenase enzymes are associated with oxo species on mononuclear iron haem and diiron non-haem platforms, respectively. Heme 139-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 23585710-2 2012 Herein we validate the use of our non-heme iron complex, Fe(PDP), as an oxidative tailoring enzyme mimic to test the proposal that this class of natural products derives from cafestol via cytochrome P-450-mediated furan oxidation. Iron 43-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-204 23585710-2 2012 Herein we validate the use of our non-heme iron complex, Fe(PDP), as an oxidative tailoring enzyme mimic to test the proposal that this class of natural products derives from cafestol via cytochrome P-450-mediated furan oxidation. Iron 57-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-204 23585710-2 2012 Herein we validate the use of our non-heme iron complex, Fe(PDP), as an oxidative tailoring enzyme mimic to test the proposal that this class of natural products derives from cafestol via cytochrome P-450-mediated furan oxidation. cafestol 175-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-204 23585710-2 2012 Herein we validate the use of our non-heme iron complex, Fe(PDP), as an oxidative tailoring enzyme mimic to test the proposal that this class of natural products derives from cafestol via cytochrome P-450-mediated furan oxidation. furan 214-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-204 22851614-4 2012 Cytochrome P450 (P450) phenotyping studies revealed that the formation of M1 and M2 were NADPH-dependent and primarily catalyzed by CYP3A4 among the studied P450 isoforms. NADP 89-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22676424-3 2012 We examined the effect rifampicin, a well-known inducer of many cytochrome P450 (CYP) enzymes and transporters, on the pharmacokinetics of intravenous and oral S-ketamine in healthy volunteers. Rifampin 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 22676424-3 2012 We examined the effect rifampicin, a well-known inducer of many cytochrome P450 (CYP) enzymes and transporters, on the pharmacokinetics of intravenous and oral S-ketamine in healthy volunteers. Rifampin 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 22738989-3 2012 In the present study, the oxidative metabolism of BDE-99 was assessed using pooled and single-donor human liver microsomes, a panel of human recombinant cytochrome P450 (CYP) enzymes, and CYP-specific antibodies. 2,2',4,4',5-brominated diphenyl ether 50-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-168 23092891-1 2012 Cytochrome P450 enzymes catalyze the biosynthesis of steroid hormones and metabolize drugs. Steroids 53-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22929863-4 2012 In this study, we show that the electrocatalytic oxidation of hydrogen peroxide on a platinum electrode generates reactive oxygen species, presumably surface-bound platinum-oxo species, which are capable of oxygen insertion reactions in analogy to oxo-ferryl radical cations in the active site of Cytochrome P450. Hydrogen Peroxide 62-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 297-312 22929863-4 2012 In this study, we show that the electrocatalytic oxidation of hydrogen peroxide on a platinum electrode generates reactive oxygen species, presumably surface-bound platinum-oxo species, which are capable of oxygen insertion reactions in analogy to oxo-ferryl radical cations in the active site of Cytochrome P450. Platinum 85-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 297-312 22929863-4 2012 In this study, we show that the electrocatalytic oxidation of hydrogen peroxide on a platinum electrode generates reactive oxygen species, presumably surface-bound platinum-oxo species, which are capable of oxygen insertion reactions in analogy to oxo-ferryl radical cations in the active site of Cytochrome P450. Reactive Oxygen Species 114-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 297-312 22929863-4 2012 In this study, we show that the electrocatalytic oxidation of hydrogen peroxide on a platinum electrode generates reactive oxygen species, presumably surface-bound platinum-oxo species, which are capable of oxygen insertion reactions in analogy to oxo-ferryl radical cations in the active site of Cytochrome P450. Platinum 164-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 297-312 22929863-4 2012 In this study, we show that the electrocatalytic oxidation of hydrogen peroxide on a platinum electrode generates reactive oxygen species, presumably surface-bound platinum-oxo species, which are capable of oxygen insertion reactions in analogy to oxo-ferryl radical cations in the active site of Cytochrome P450. Oxygen 123-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 297-312 22927483-3 2012 Sorafenib metabolism was evaluated in vitro at 10 mumol/L using a panel of purified human cytochrome P450 (CYP) enzymes. Sorafenib 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 22927483-3 2012 Sorafenib metabolism was evaluated in vitro at 10 mumol/L using a panel of purified human cytochrome P450 (CYP) enzymes. Sorafenib 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-110 22828666-7 2012 At a low dose, 5 mug/ml (15 muM), carnosic acid activated the expression of 3 genes, induced through the presence of antioxidant response elements, including genes involved in glutathione biosynthesis (CYP4F3, GCLC) and transport (SLC7A11). salvin 34-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-208 22828666-7 2012 At a low dose, 5 mug/ml (15 muM), carnosic acid activated the expression of 3 genes, induced through the presence of antioxidant response elements, including genes involved in glutathione biosynthesis (CYP4F3, GCLC) and transport (SLC7A11). Glutathione 176-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-208 22738989-3 2012 In the present study, the oxidative metabolism of BDE-99 was assessed using pooled and single-donor human liver microsomes, a panel of human recombinant cytochrome P450 (CYP) enzymes, and CYP-specific antibodies. 2,2',4,4',5-brominated diphenyl ether 50-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-173 22738989-3 2012 In the present study, the oxidative metabolism of BDE-99 was assessed using pooled and single-donor human liver microsomes, a panel of human recombinant cytochrome P450 (CYP) enzymes, and CYP-specific antibodies. 2,2',4,4',5-brominated diphenyl ether 50-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-191 22621785-3 2012 Epoxyeicosatrienoic acids (EETs), products of cytochrome P-450 epoxidation of arachidonic acid, have vasculoprotective and anti-inflammatory effects including inhibition of platelet activation, cell migration, and adhesion. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 22639088-0 2012 Defluorination of 4-fluorophenol by cytochrome P450(BM3)-F87G: activation by long chain fatty aldehydes. 4-fluorophenol 18-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-55 22639088-0 2012 Defluorination of 4-fluorophenol by cytochrome P450(BM3)-F87G: activation by long chain fatty aldehydes. long chain fatty aldehydes 77-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-55 22639088-1 2012 Cytochrome P450(BM3)-F87G catalyzed the oxidative defluorination of 4-fluorophenol, followed by reduction of the resulting benzoquinone to hydroquinone via the NADPH P450-reductase activity of the enzyme. 4-fluorophenol 68-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-19 22639088-1 2012 Cytochrome P450(BM3)-F87G catalyzed the oxidative defluorination of 4-fluorophenol, followed by reduction of the resulting benzoquinone to hydroquinone via the NADPH P450-reductase activity of the enzyme. quinone 123-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-19 22639088-1 2012 Cytochrome P450(BM3)-F87G catalyzed the oxidative defluorination of 4-fluorophenol, followed by reduction of the resulting benzoquinone to hydroquinone via the NADPH P450-reductase activity of the enzyme. hydroquinone 139-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-19 22411763-1 2012 Previously, our laboratory reported that cyclic peptide prodrugs of the opioid peptide H-Tyr-D-Ala-Gly-Phe-D-Leu-OH (DADLE) are metabolized by cytochrome P450 (CYP450) enzymes, which limits their systemic exposure after oral dosing to animals. DADLE 87-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-158 22411763-1 2012 Previously, our laboratory reported that cyclic peptide prodrugs of the opioid peptide H-Tyr-D-Ala-Gly-Phe-D-Leu-OH (DADLE) are metabolized by cytochrome P450 (CYP450) enzymes, which limits their systemic exposure after oral dosing to animals. DADLE 87-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-166 22411763-1 2012 Previously, our laboratory reported that cyclic peptide prodrugs of the opioid peptide H-Tyr-D-Ala-Gly-Phe-D-Leu-OH (DADLE) are metabolized by cytochrome P450 (CYP450) enzymes, which limits their systemic exposure after oral dosing to animals. DADLE 117-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-158 22411763-1 2012 Previously, our laboratory reported that cyclic peptide prodrugs of the opioid peptide H-Tyr-D-Ala-Gly-Phe-D-Leu-OH (DADLE) are metabolized by cytochrome P450 (CYP450) enzymes, which limits their systemic exposure after oral dosing to animals. DADLE 117-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-166 22634049-4 2012 By both a pharmacological and a siRNA approaches, we demonstrated that recruitment of the Pregnane X Receptor (PXR) was required to mediate CYP4F3 induction by lovastatin. Lovastatin 160-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-146 22634049-5 2012 Furthermore, the CYP4F3 gene promoter was transcriptionally activated by PXR, and responded to lovastatin. Lovastatin 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 22634049-6 2012 Finally, the expression of fatty acid-responsive genes was increased in response to the statin or 20-HETE in a CYP4F3-dependent way. Fatty Acids 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-117 22634049-7 2012 We propose that metabolites produced by CYP4F3 could modulate lipid metabolism in response to lovastatin. Lovastatin 94-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-46 22808947-1 2012 Although pharmaceutical excipients are supposed to be pharmacologically inactive, solubilizing agents like Cremophor EL have been shown to interact with cytochrome P450 (CYP)-dependent drug metabolism as well as efflux transporters such as P-glycoprotein (ABCB1) and multidrug resistance associated protein 2 (ABCC2). cremophor EL 107-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-168 22808947-1 2012 Although pharmaceutical excipients are supposed to be pharmacologically inactive, solubilizing agents like Cremophor EL have been shown to interact with cytochrome P450 (CYP)-dependent drug metabolism as well as efflux transporters such as P-glycoprotein (ABCB1) and multidrug resistance associated protein 2 (ABCC2). cremophor EL 107-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-173 22773874-2 2012 We demonstrated that, like testosterone, dihydrotestosterone can be oxidized by human cytochrome P450 (P450) 19A1, the steroid aromatase. Testosterone 27-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-113 22773874-2 2012 We demonstrated that, like testosterone, dihydrotestosterone can be oxidized by human cytochrome P450 (P450) 19A1, the steroid aromatase. Dihydrotestosterone 41-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-113 22610824-0 2012 Metabolic-intermediate complex formation with cytochrome P450: theoretical studies in elucidating the reaction pathway for the generation of reactive nitroso intermediate. reactive 141-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 22610824-0 2012 Metabolic-intermediate complex formation with cytochrome P450: theoretical studies in elucidating the reaction pathway for the generation of reactive nitroso intermediate. nitroso intermediate 150-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 22610824-7 2012 The study was performed using Cpd I [iron (IV-oxo] heme porphine with SH(-) as the axial ligand) to represent the catalytic domain of CYP, simulating the biotransformation process. iron (iv-oxo] heme porphine 37-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-137 22621785-3 2012 Epoxyeicosatrienoic acids (EETs), products of cytochrome P-450 epoxidation of arachidonic acid, have vasculoprotective and anti-inflammatory effects including inhibition of platelet activation, cell migration, and adhesion. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 22621785-3 2012 Epoxyeicosatrienoic acids (EETs), products of cytochrome P-450 epoxidation of arachidonic acid, have vasculoprotective and anti-inflammatory effects including inhibition of platelet activation, cell migration, and adhesion. Arachidonic Acid 78-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 22173545-0 2012 Hypertension, cardiovascular risk and polymorphisms in genes controlling the cytochrome P450 pathway of arachidonic acid: A sex-specific relation? Arachidonic Acid 104-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 22101002-1 2012 The thick ascending limb of Henle"s loop (TAL) is capable of metabolizing arachidonic acid (AA) by cytochrome P450 (CYP450) and cyclooxygenase (COX) pathways and has been identified as a nephron segment that contributes to salt-sensitive hypertension. Arachidonic Acid 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-114 22686547-14 2012 Saxagliptin and, to a lesser extent, sitagliptin are largely metabolized by hepatic cytochrome P450 (CYP) 3A4 and 3A5 isoforms. saxagliptin 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 22686547-14 2012 Saxagliptin and, to a lesser extent, sitagliptin are largely metabolized by hepatic cytochrome P450 (CYP) 3A4 and 3A5 isoforms. saxagliptin 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 22686547-14 2012 Saxagliptin and, to a lesser extent, sitagliptin are largely metabolized by hepatic cytochrome P450 (CYP) 3A4 and 3A5 isoforms. Sitagliptin Phosphate 37-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 22686547-14 2012 Saxagliptin and, to a lesser extent, sitagliptin are largely metabolized by hepatic cytochrome P450 (CYP) 3A4 and 3A5 isoforms. Sitagliptin Phosphate 37-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 22788578-7 2012 In this review, the known in vitro and in vivo effects of resveratrol on various cytochrome P450 (CYP) isoenzymes are summarized. Resveratrol 58-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 22788578-7 2012 In this review, the known in vitro and in vivo effects of resveratrol on various cytochrome P450 (CYP) isoenzymes are summarized. Resveratrol 58-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 22101002-1 2012 The thick ascending limb of Henle"s loop (TAL) is capable of metabolizing arachidonic acid (AA) by cytochrome P450 (CYP450) and cyclooxygenase (COX) pathways and has been identified as a nephron segment that contributes to salt-sensitive hypertension. Arachidonic Acid 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-122 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. Arachidonic Acid 45-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. Arachidonic Acid 45-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. epoxyeicosatrienoic acids 70-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. epoxyeicosatrienoic acids 70-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. eets 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. eets 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 107-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 107-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 140-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 140-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. Sodium 206-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22173545-2 2012 Cytochrome P450 (CYP)-derived metabolites of arachidonic acid such as epoxyeicosatrienoic acids (EETs) and 20-hydroxyeicosatetraenoic acid (20-HETE), active on vascular tone, endothelial function and renal sodium reapportion, have been identified as candidate mediators in the development of hypertension in several animal models, with remarkable sex-specific effect. Sodium 206-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 22513143-2 2012 Sorafenib also undergoes cytochrome P450 (CYP)-dependent biotransformation to the N-oxide and other metabolites. Sorafenib 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 22513143-2 2012 Sorafenib also undergoes cytochrome P450 (CYP)-dependent biotransformation to the N-oxide and other metabolites. Sorafenib 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 22513143-2 2012 Sorafenib also undergoes cytochrome P450 (CYP)-dependent biotransformation to the N-oxide and other metabolites. n-oxide 82-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 22513143-2 2012 Sorafenib also undergoes cytochrome P450 (CYP)-dependent biotransformation to the N-oxide and other metabolites. n-oxide 82-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 22513143-4 2012 In the present study CYP-mediated pathways of sorafenib oxidation in human liver were evaluated. Sorafenib 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 22513143-7 2012 In human hepatic microsomes metabolite formation was correlated with CYP3A4-mediated midazolam 1"-hydroxylation, but not with other CYP-specific substrate oxidations. Midazolam 85-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 22541353-4 2012 High CYP expression and activities could be further enhanced by culturing the induced HDFs either as spheroids or into several kinds of scaffolds, particularly the tri-copolymer scaffold composed of gelatin, chondroitin and hyaluronan. tri-copolymer 164-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-8 22541353-4 2012 High CYP expression and activities could be further enhanced by culturing the induced HDFs either as spheroids or into several kinds of scaffolds, particularly the tri-copolymer scaffold composed of gelatin, chondroitin and hyaluronan. Chondroitin 208-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-8 22541353-4 2012 High CYP expression and activities could be further enhanced by culturing the induced HDFs either as spheroids or into several kinds of scaffolds, particularly the tri-copolymer scaffold composed of gelatin, chondroitin and hyaluronan. Hyaluronic Acid 224-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-8 22513183-2 2012 This review focuses on the therapeutic promise of dihydroquercetin in major disease states such as cancer, cardiovascular disease and liver disease by reviewing the proposed mechanism(s) of action, including the activation of the antioxidant response element (ARE) and detoxifying phase II enzymes, inhibition of cytochrome P(450) and fatty acid synthase in carcinogenesis. taxifolin 50-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 313-330 22233204-12 2012 RESULTS: In vitro studies demonstrated low potential for CYP inhibition (IC(50) estimates tofacitinib > 30 microm), CYP3A4 mRNA induction (observed at tofacitinib concentrations >= 25 microm) and no effect on enzymatic activity of CYP substrates. tofacitinib 90-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 22568693-6 2012 Expression of the 3435T/T genetic variant encoding the MDR1 gene for the P-glycoprotein efflux transporter results in a significantly reduced maximum drug concentration and area under the plasma concentration-time curve as intestinal absorption of clopidogrel is reduced; and the expression of the mutant *2 allele of CYP2C19 results in similar pharmacokinetic effects as the two cytochrome P450 (CYP)-mediated steps required for the production of the active metabolite of clopidogrel are impaired. Clopidogrel 248-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 380-395 22568693-6 2012 Expression of the 3435T/T genetic variant encoding the MDR1 gene for the P-glycoprotein efflux transporter results in a significantly reduced maximum drug concentration and area under the plasma concentration-time curve as intestinal absorption of clopidogrel is reduced; and the expression of the mutant *2 allele of CYP2C19 results in similar pharmacokinetic effects as the two cytochrome P450 (CYP)-mediated steps required for the production of the active metabolite of clopidogrel are impaired. Clopidogrel 248-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 318-321 22568693-6 2012 Expression of the 3435T/T genetic variant encoding the MDR1 gene for the P-glycoprotein efflux transporter results in a significantly reduced maximum drug concentration and area under the plasma concentration-time curve as intestinal absorption of clopidogrel is reduced; and the expression of the mutant *2 allele of CYP2C19 results in similar pharmacokinetic effects as the two cytochrome P450 (CYP)-mediated steps required for the production of the active metabolite of clopidogrel are impaired. Clopidogrel 473-484 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 380-395 22568693-6 2012 Expression of the 3435T/T genetic variant encoding the MDR1 gene for the P-glycoprotein efflux transporter results in a significantly reduced maximum drug concentration and area under the plasma concentration-time curve as intestinal absorption of clopidogrel is reduced; and the expression of the mutant *2 allele of CYP2C19 results in similar pharmacokinetic effects as the two cytochrome P450 (CYP)-mediated steps required for the production of the active metabolite of clopidogrel are impaired. Clopidogrel 473-484 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 318-321 22568693-9 2012 Neither appears to be affected by the same genetic polymorphisms as clopidogrel; prasugrel requires only a single CYP-mediated step to produce its active metabolite and ticagrelor is not a pro-drug. Prasugrel Hydrochloride 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 22692316-0 2012 [Metabolic detoxification of bakuchiol is mediated by cytochrome P450 enzymes in human liver microsomes]. bakuchiol 29-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 22692316-1 2012 OBJECTIVE: To analyze cytochrome P450 (CYP) phenotyping for bakuchiol metabolism and study the mechanism of detoxification of bakuchiol by human liver microsomes (HLM) in vitro. bakuchiol 60-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 22692316-1 2012 OBJECTIVE: To analyze cytochrome P450 (CYP) phenotyping for bakuchiol metabolism and study the mechanism of detoxification of bakuchiol by human liver microsomes (HLM) in vitro. bakuchiol 60-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 22692316-2 2012 METHODS: The CYP phenotyping for bakuchiol metabolism was determined using HLM combined with CYP specific inhibitors and recombinant human CYP isoforms. bakuchiol 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 22692316-2 2012 METHODS: The CYP phenotyping for bakuchiol metabolism was determined using HLM combined with CYP specific inhibitors and recombinant human CYP isoforms. bakuchiol 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 22692316-2 2012 METHODS: The CYP phenotyping for bakuchiol metabolism was determined using HLM combined with CYP specific inhibitors and recombinant human CYP isoforms. bakuchiol 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 22692316-3 2012 The relative activities of CYP isoforms were determined by analyzing the formation of the substrate metabolites using HPLC-MS/MS, in presence or absence of 1-aminobenzotriazole (ABT) which was CYP enzymes" broad spectrum inhibitor. 1-aminobenzotriazole 157-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 22692316-3 2012 The relative activities of CYP isoforms were determined by analyzing the formation of the substrate metabolites using HPLC-MS/MS, in presence or absence of 1-aminobenzotriazole (ABT) which was CYP enzymes" broad spectrum inhibitor. 1-aminobenzotriazole 157-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-197 22692316-3 2012 The relative activities of CYP isoforms were determined by analyzing the formation of the substrate metabolites using HPLC-MS/MS, in presence or absence of 1-aminobenzotriazole (ABT) which was CYP enzymes" broad spectrum inhibitor. 1-aminobenzotriazole 179-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 22692316-3 2012 The relative activities of CYP isoforms were determined by analyzing the formation of the substrate metabolites using HPLC-MS/MS, in presence or absence of 1-aminobenzotriazole (ABT) which was CYP enzymes" broad spectrum inhibitor. 1-aminobenzotriazole 179-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-197 22692316-5 2012 The effects of CYP enzymes on the nephrotoxicity of bakuchiol were investigated using human kidney-2(HK-2) by MTT assay, in presence or absence of ABT. bakuchiol 52-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 22692316-10 2012 CONCLUSION: The mechanism of metabolic detoxification of bakuchiol by HLM is associated with bakuchiol metabolism by CYP enzymes to form non toxic or lower toxic metabolites. bakuchiol 57-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 22692316-10 2012 CONCLUSION: The mechanism of metabolic detoxification of bakuchiol by HLM is associated with bakuchiol metabolism by CYP enzymes to form non toxic or lower toxic metabolites. bakuchiol 94-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 22233204-2 2012 Non-renal elimination accounts for 70% of the total clearance of tofacitinib and the metabolism is primarily mediated by cytochrome P450 (CYP) 3A4. tofacitinib 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-136 22233204-2 2012 Non-renal elimination accounts for 70% of the total clearance of tofacitinib and the metabolism is primarily mediated by cytochrome P450 (CYP) 3A4. tofacitinib 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 22617226-0 2012 Effects of CYP induction by rifampicin on tamoxifen exposure. Rifampin 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-14 22617226-0 2012 Effects of CYP induction by rifampicin on tamoxifen exposure. Tamoxifen 42-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-14 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Rifampin 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Rifampin 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Tamoxifen 100-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Tamoxifen 100-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Rifampin 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Rifampin 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 22617226-3 2012 We evaluated the effects of cytochrome P450 (CYP) induction by rifampicin on the exposure levels of tamoxifen and its metabolites and found that coadministration of rifampicin resulted in markedly reduced (up to 86%, P <= 0.040) concentrations of tamoxifen and its metabolites. Tamoxifen 250-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 22617226-5 2012 Similar drug-drug interactions may exist between tamoxifen and other strong CYP inducers. Tamoxifen 49-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 22568886-4 2012 Several applications in drug discovery studies are discussed and the use of HepaRG, as a human relevant predictive in vitro CYP induction model, is described. heparg 76-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 22426923-3 2012 PATIENTS AND METHODS: 79 SNPs in CYP450, whose minor allele frequency were >= 10%, were genotyped in 69 MBC patients who were treated with docetaxel plus capecitabine. Docetaxel 142-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-39 22426923-3 2012 PATIENTS AND METHODS: 79 SNPs in CYP450, whose minor allele frequency were >= 10%, were genotyped in 69 MBC patients who were treated with docetaxel plus capecitabine. Capecitabine 157-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-39 22503544-0 2012 Evaluation of cytochrome P450-derived eicosanoids in humans with stable atherosclerotic cardiovascular disease. Eicosanoids 38-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 22503544-1 2012 OBJECTIVE: Preclinical and genetic epidemiologic studies suggest that modulating cytochrome P450 (CYP)-mediated arachidonic acid metabolism may have therapeutic utility in the management of coronary artery disease (CAD). Arachidonic Acid 112-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 22503544-1 2012 OBJECTIVE: Preclinical and genetic epidemiologic studies suggest that modulating cytochrome P450 (CYP)-mediated arachidonic acid metabolism may have therapeutic utility in the management of coronary artery disease (CAD). Arachidonic Acid 112-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 22503544-2 2012 However, predictors of inter-individual variation in CYP-derived eicosanoid metabolites in CAD patients have not been evaluated to date. Eicosanoids 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 22503544-9 2012 CONCLUSIONS: Collectively, these findings suggest that CYP-mediated eicosanoid metabolism is dysregulated in certain subsets of CAD patients, and demonstrate that biomarkers of CYP epoxygenase and sEH, but not CYP omega-hydroxylase, metabolism are altered in stable CAD patients relative to healthy individuals. Eicosanoids 68-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. 6-oh-3mi 233-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 22554311-10 2012 DISCUSSION/CONCLUSION: The results suggest that fomepizole induces its own metabolism via cytochrome P-450, leading to enhanced fomepizole elimination and 4-CP excretion. Fomepizole 48-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 22554311-10 2012 DISCUSSION/CONCLUSION: The results suggest that fomepizole induces its own metabolism via cytochrome P-450, leading to enhanced fomepizole elimination and 4-CP excretion. Fomepizole 128-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 22554311-10 2012 DISCUSSION/CONCLUSION: The results suggest that fomepizole induces its own metabolism via cytochrome P-450, leading to enhanced fomepizole elimination and 4-CP excretion. 4-carboxypyrazole 155-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 22493087-2 2012 Recent evidence suggests that epoxyeicosatrienoic acid (EET), which is derived from arachidonic acid by cytochrome p450 (CYP) epoxygenase, has an essential role in PAH. epoxyeicosatrienoic acid 30-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 22493087-2 2012 Recent evidence suggests that epoxyeicosatrienoic acid (EET), which is derived from arachidonic acid by cytochrome p450 (CYP) epoxygenase, has an essential role in PAH. epoxyeicosatrienoic acid 30-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 22493087-2 2012 Recent evidence suggests that epoxyeicosatrienoic acid (EET), which is derived from arachidonic acid by cytochrome p450 (CYP) epoxygenase, has an essential role in PAH. EET 56-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 22493087-2 2012 Recent evidence suggests that epoxyeicosatrienoic acid (EET), which is derived from arachidonic acid by cytochrome p450 (CYP) epoxygenase, has an essential role in PAH. EET 56-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 22493087-2 2012 Recent evidence suggests that epoxyeicosatrienoic acid (EET), which is derived from arachidonic acid by cytochrome p450 (CYP) epoxygenase, has an essential role in PAH. Arachidonic Acid 84-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 22493087-2 2012 Recent evidence suggests that epoxyeicosatrienoic acid (EET), which is derived from arachidonic acid by cytochrome p450 (CYP) epoxygenase, has an essential role in PAH. Arachidonic Acid 84-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 22616812-2 2012 Further, clinically significant interactions with inhibitors of cytochrome P450 (CYP) have previously been reported for drugs of this therapeutic group, such as terfenadine and astemizole, indicating the possibility of drug-drug interactions involving agents that share the same metabolic pathway. Terfenadine 161-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 22616812-2 2012 Further, clinically significant interactions with inhibitors of cytochrome P450 (CYP) have previously been reported for drugs of this therapeutic group, such as terfenadine and astemizole, indicating the possibility of drug-drug interactions involving agents that share the same metabolic pathway. Terfenadine 161-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 22616812-2 2012 Further, clinically significant interactions with inhibitors of cytochrome P450 (CYP) have previously been reported for drugs of this therapeutic group, such as terfenadine and astemizole, indicating the possibility of drug-drug interactions involving agents that share the same metabolic pathway. Astemizole 177-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 22616812-2 2012 Further, clinically significant interactions with inhibitors of cytochrome P450 (CYP) have previously been reported for drugs of this therapeutic group, such as terfenadine and astemizole, indicating the possibility of drug-drug interactions involving agents that share the same metabolic pathway. Astemizole 177-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 22616812-3 2012 The aim of this article was to review the preclinical testing of a new antihistamine (i.e., bilastine) in terms of its biotransformation in various animal species, including humans, and to evaluate its potential for possible drug-drug interactions involving the CYP system. bilastine 92-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-265 22627182-1 2012 OBJECTIVES: Pitavastatin, a statin recently approved in the United States, has a potential benefit of reduced risk of cytochrome P450 (CYP)-mediated drug-drug interaction due to minimal metabolism by the CYP system. pitavastatin 12-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 22627182-1 2012 OBJECTIVES: Pitavastatin, a statin recently approved in the United States, has a potential benefit of reduced risk of cytochrome P450 (CYP)-mediated drug-drug interaction due to minimal metabolism by the CYP system. pitavastatin 12-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 22627182-1 2012 OBJECTIVES: Pitavastatin, a statin recently approved in the United States, has a potential benefit of reduced risk of cytochrome P450 (CYP)-mediated drug-drug interaction due to minimal metabolism by the CYP system. pitavastatin 12-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-207 22558931-0 2012 The roles of different porcine cytochrome P450 enzymes and cytochrome b5A in skatole metabolism. Skatole 77-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. Skatole 19-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. Skatole 19-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-148 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. 3-methyl-1H-indol-6-ol 207-231 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. 3-methyl-1H-indol-6-ol 207-231 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-148 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. 6-oh-3mi 233-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-148 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. 3-methyl-1H-indol-6-ol 243-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. 3-methyl-1H-indol-6-ol 243-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-148 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. Skatole 252-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 22558931-2 2012 The key enzymes in skatole metabolism are thought to be cytochrome P450 2E1 (CYP2E1) and cytochrome 2A (CYP2A); however, the cytochrome P450 (CYP450) isoform responsible for the production of the metabolite 6-hydroxy-3-methylindole (6-OH-3MI, 6-hydroxyskatole), which is thought to be involved in the clearance of skatole, has not been established conclusively. Skatole 252-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-148 22558931-3 2012 The aim of this study was to characterize the role of porcine CYP450s in skatole metabolism by expressing them individually in the human embryonic kidney HEK293-FT cell line. Skatole 73-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-68 22558931-9 2012 Further information regarding the abundance of different CYP450 isoforms is required to fully understand their contribution to skatole metabolism in vivo in the pig. Skatole 127-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-63 22051186-4 2012 We have determined how a related set of three human CYP enzymes bind and interact with a common inhibitor, the muscarinic receptor agonist drug pilocarpine. Pilocarpine 144-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 28509155-1 2012 It is well known that miconazole inhibits cytochrome P450 (CYP). Miconazole 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 28509155-1 2012 It is well known that miconazole inhibits cytochrome P450 (CYP). Miconazole 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 22471442-0 2012 Computational toxicological investigation on the mechanism and pathways of xenobiotics metabolized by cytochrome P450: a case of BDE-47. 2,2',4,4'-tetrabromodiphenyl ether 129-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-117 22483397-2 2012 CYP isoform specific substrates and their metabolites of CYP1A2 (caffeine), CYP2C9 (losartan), CYP2C19 (omeprazole), CYP2D6 (dextromethorphan) and CYP3A (midazolam) were all simultaneously analyzed using LC-MS/MS after administration of a mixture of five drugs (i.e., a "cocktail approach") to healthy volunteers. Losartan 84-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21751216-6 2012 Moreover, cells exposed to the combination of OSM, DEX, and HGF gradually featured highly differentiated hepatic functions, including ALB secretion, glycogen storage, urea production, and cytochrome P450 (CYP) activity. Dexamethasone 51-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-203 21751216-6 2012 Moreover, cells exposed to the combination of OSM, DEX, and HGF gradually featured highly differentiated hepatic functions, including ALB secretion, glycogen storage, urea production, and cytochrome P450 (CYP) activity. Dexamethasone 51-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-208 22483397-2 2012 CYP isoform specific substrates and their metabolites of CYP1A2 (caffeine), CYP2C9 (losartan), CYP2C19 (omeprazole), CYP2D6 (dextromethorphan) and CYP3A (midazolam) were all simultaneously analyzed using LC-MS/MS after administration of a mixture of five drugs (i.e., a "cocktail approach") to healthy volunteers. Caffeine 65-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 22330698-7 2012 In multivariate regression models a significant positive correlation between serum vitamin D levels and the expression of vitamin D-regulated targets, cytochrome P450, family 24, subfamily a (cyp24a) expression by PBMCs (P = .0084, pediatric asthma group only) and serum LL-37 levels (P = .0006 in the pediatric group but P = .0067 in the adult asthma group), was found. Vitamin D 83-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-177 22483397-2 2012 CYP isoform specific substrates and their metabolites of CYP1A2 (caffeine), CYP2C9 (losartan), CYP2C19 (omeprazole), CYP2D6 (dextromethorphan) and CYP3A (midazolam) were all simultaneously analyzed using LC-MS/MS after administration of a mixture of five drugs (i.e., a "cocktail approach") to healthy volunteers. Omeprazole 104-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 22483397-2 2012 CYP isoform specific substrates and their metabolites of CYP1A2 (caffeine), CYP2C9 (losartan), CYP2C19 (omeprazole), CYP2D6 (dextromethorphan) and CYP3A (midazolam) were all simultaneously analyzed using LC-MS/MS after administration of a mixture of five drugs (i.e., a "cocktail approach") to healthy volunteers. Dextromethorphan 125-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 22483397-2 2012 CYP isoform specific substrates and their metabolites of CYP1A2 (caffeine), CYP2C9 (losartan), CYP2C19 (omeprazole), CYP2D6 (dextromethorphan) and CYP3A (midazolam) were all simultaneously analyzed using LC-MS/MS after administration of a mixture of five drugs (i.e., a "cocktail approach") to healthy volunteers. Midazolam 154-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 22266842-0 2012 Identification of human cytochrome P450 and flavin-containing monooxygenase enzymes involved in the metabolism of lorcaserin, a novel selective human 5-hydroxytryptamine 2C agonist. lorcaserin 114-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 22706230-3 2012 Apart from the omega-hydroxylation of leukotriene B4 and prostaglandins, CYP4F3 is the main catalyst in the oxidation of fatty acid epoxides. Leukotriene B4 38-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-79 22706230-3 2012 Apart from the omega-hydroxylation of leukotriene B4 and prostaglandins, CYP4F3 is the main catalyst in the oxidation of fatty acid epoxides. fatty acid epoxides 121-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-79 22252457-0 2012 Comment on "binding free energies of inhibitors to iron porphyrin complex as a model for cytochrome P450". iron porphyrin 51-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 22317799-4 2012 Ideally, such an assay would use a single PCR reaction and, without further processing, a single microchip electrophoresis (ME) run to determine the 3 single-nucleotide polymorphisms (SNPs) affecting warfarin sensitivity [i.e., CYP2C9 (cytochrome P450, family 2, subfamily C, polypeptide 9) *2, CYP2C9 *3, and the VKORC1 (vitamin K epoxide reductase complex 1) A/B haplotype]. Warfarin 200-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 236-261 22510212-0 2012 Role of perferryl-oxo oxidant in alkane hydroxylation catalyzed by cytochrome P450: a hybrid density functional study. Alkanes 33-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 22113809-0 2012 Binding free energies of inhibitors to iron porphyrin complex as a model for Cytochrome P450. iron porphyrin 39-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 22266842-0 2012 Identification of human cytochrome P450 and flavin-containing monooxygenase enzymes involved in the metabolism of lorcaserin, a novel selective human 5-hydroxytryptamine 2C agonist. Serotonin 150-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 22422635-0 2012 Pharmacokinetic drug interactions between clobazam and drugs metabolized by cytochrome P450 isoenzymes. Clobazam 42-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 22589111-3 2012 Kinetics of clopidogrel metabolism is driven by enzymatic activity of the Cytochrome P450 system. Clopidogrel 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 22366455-5 2012 Absence of cytochrome P450, family 26, subfamily b, polypeptide 1 (Cyp26b1), a retinoic-acid-degrading enzyme, results in aberrant epidermal differentiation and filaggrin expression, defective cornified envelopes and skin barrier formation, in conjunction with peridermal retention. Tretinoin 79-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-50 22422635-1 2012 STUDY OBJECTIVE: To investigate potential drug-drug interactions between clobazam and cytochrome P450 (CYP) isoenzyme substrates, inhibitors, and inducers. Clobazam 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 22422635-1 2012 STUDY OBJECTIVE: To investigate potential drug-drug interactions between clobazam and cytochrome P450 (CYP) isoenzyme substrates, inhibitors, and inducers. Clobazam 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 22330256-2 2012 Recent investigations at our lab have shown that the metabolites generated through cytochrome P(450)-dependent metabolic reactions are responsible for hemotoxic effects of primaquine, which could be monitored with accumulation of methemoglobin and increased oxidative stress. Primaquine 172-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-100 21415279-0 2012 Influence of different proton pump inhibitors on activity of cytochrome P450 assessed by [(13)C]-aminopyrine breath test. Aminopyrine 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 21415279-1 2012 Aminopyrine is metabolized by cytochrome P450 (CYP) in the liver. Aminopyrine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 21415279-1 2012 Aminopyrine is metabolized by cytochrome P450 (CYP) in the liver. Aminopyrine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 21415279-6 2012 Omeprazole and lansoprazole at the standard doses inhibit CYP activity but rabeprazole does not, whereas high-dose omeprazole seems to induce CYPs. Omeprazole 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 21415279-6 2012 Omeprazole and lansoprazole at the standard doses inhibit CYP activity but rabeprazole does not, whereas high-dose omeprazole seems to induce CYPs. Lansoprazole 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 22022918-3 2012 In the present study, several in vitro approaches were used to identify the cytochrome P450 (CYP) enzymes responsible for ilaprazole sulfone formation. ilaprazole sulfone 122-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 22022918-3 2012 In the present study, several in vitro approaches were used to identify the cytochrome P450 (CYP) enzymes responsible for ilaprazole sulfone formation. ilaprazole sulfone 122-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 20740300-1 2012 OBJECTIVE: Axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptors 1, 2, and 3, is metabolized primarily by cytochrome P450 (CYP) 3A with minor contributions from CYP1A2, CYP2C19, and glucuronidation. Axitinib 11-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-178 22155354-4 2012 The AhR has particular importance in the lung and is most commonly associated with the up-regulation of cytochrome P-450 (CYP)-mediated metabolism of benzo[a]pyrene (B[a]P) to reactive intermediates. Benzo(a)pyrene 150-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 22155354-4 2012 The AhR has particular importance in the lung and is most commonly associated with the up-regulation of cytochrome P-450 (CYP)-mediated metabolism of benzo[a]pyrene (B[a]P) to reactive intermediates. Benzo(a)pyrene 150-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 26596595-0 2012 Development of Accurate DFT Methods for Computing Redox Potentials of Transition Metal Complexes: Results for Model Complexes and Application to Cytochrome P450. Metals 81-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 26596595-8 2012 Application of the LOC scheme to the rate-determining hydrogen atom transfer step in substrate hydroxylation by cytochrome P450 shows that this approach is able to correct the B3LYP barriers in comparison to recent kinetics experiments. Hydrogen 54-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-127 22050110-1 2012 The steroid and xenobiotic receptor (SXR) regulates cytochrome P450 (CYP) enzymes, which are key inactivators of testosterone in the liver and prostate. Testosterone 113-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 22050110-1 2012 The steroid and xenobiotic receptor (SXR) regulates cytochrome P450 (CYP) enzymes, which are key inactivators of testosterone in the liver and prostate. Testosterone 113-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifampin 126-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 20740300-1 2012 OBJECTIVE: Axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptors 1, 2, and 3, is metabolized primarily by cytochrome P450 (CYP) 3A with minor contributions from CYP1A2, CYP2C19, and glucuronidation. Axitinib 11-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifampin 126-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-95 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifampin 138-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 20740300-1 2012 OBJECTIVE: Axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptors 1, 2, and 3, is metabolized primarily by cytochrome P450 (CYP) 3A with minor contributions from CYP1A2, CYP2C19, and glucuronidation. Axitinib 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-178 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifampin 138-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-95 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). rifapentine 144-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 20740300-1 2012 OBJECTIVE: Axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptors 1, 2, and 3, is metabolized primarily by cytochrome P450 (CYP) 3A with minor contributions from CYP1A2, CYP2C19, and glucuronidation. Axitinib 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). rifapentine 144-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-95 20740300-2 2012 Co-administration with CYP inhibitors may increase systemic exposure to axitinib and alter its safety profile. Axitinib 72-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifabutin 166-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 21899994-5 2012 DG extract increased the production of cytochrome P-450 (CYP)-dependent reactive oxygen species (ROS) in H9c2 cardiomyocytes, which was accompanied by the concomitant activation of ERK1/2 and PKCepsilon. Reactive Oxygen Species 72-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 22236925-1 2012 A study was carried out to establish the relative contribution of human cytochrome P450 (CYP450) enzymes in the metabolism of rifampicin (RMP), rifapentine (RPT) and rifabutin (RFB). Rifabutin 166-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-95 22197672-4 2012 First is accessibility of each substrate atom to the oxygenated Fe atom of heme in a CYP protein, and the other is the oxidative reactivity of each substrate atom. Iron 64-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 22197672-4 2012 First is accessibility of each substrate atom to the oxygenated Fe atom of heme in a CYP protein, and the other is the oxidative reactivity of each substrate atom. Heme 75-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 21899994-5 2012 DG extract increased the production of cytochrome P-450 (CYP)-dependent reactive oxygen species (ROS) in H9c2 cardiomyocytes, which was accompanied by the concomitant activation of ERK1/2 and PKCepsilon. Reactive Oxygen Species 72-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 21899994-5 2012 DG extract increased the production of cytochrome P-450 (CYP)-dependent reactive oxygen species (ROS) in H9c2 cardiomyocytes, which was accompanied by the concomitant activation of ERK1/2 and PKCepsilon. Reactive Oxygen Species 97-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 21899994-5 2012 DG extract increased the production of cytochrome P-450 (CYP)-dependent reactive oxygen species (ROS) in H9c2 cardiomyocytes, which was accompanied by the concomitant activation of ERK1/2 and PKCepsilon. Reactive Oxygen Species 97-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 21899994-8 2012 In conclusion, DG treatment activated both ERK/Nrf2 and PKCepsilon pathways, presumably by ROS arising from CYP-catalyzed processes, with resultant inhibition of hypoxia/reoxygenation-induced apoptosis immediately after DG treatment or even after an extended time interval following DG treatment. Reactive Oxygen Species 91-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 22687469-1 2012 Mammalian cytochrome P450 (CYP) comprise a large group of enzymes that play many important roles in the biosynthesis of steroid hormones and vitamins. Steroids 120-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 23013331-6 2012 20-Hydroxyeicosatetraenoic acid (20-HETE) is a vasoconstrictor omega-hydroxylation product of arachidonic acid that is produced by cytochrome P450 (CYP) enzymes, mainly by CYP4A and CYP4F isoforms. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 0-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 23013331-6 2012 20-Hydroxyeicosatetraenoic acid (20-HETE) is a vasoconstrictor omega-hydroxylation product of arachidonic acid that is produced by cytochrome P450 (CYP) enzymes, mainly by CYP4A and CYP4F isoforms. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 0-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 23013331-6 2012 20-Hydroxyeicosatetraenoic acid (20-HETE) is a vasoconstrictor omega-hydroxylation product of arachidonic acid that is produced by cytochrome P450 (CYP) enzymes, mainly by CYP4A and CYP4F isoforms. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 33-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 23013331-6 2012 20-Hydroxyeicosatetraenoic acid (20-HETE) is a vasoconstrictor omega-hydroxylation product of arachidonic acid that is produced by cytochrome P450 (CYP) enzymes, mainly by CYP4A and CYP4F isoforms. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 33-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 23013331-6 2012 20-Hydroxyeicosatetraenoic acid (20-HETE) is a vasoconstrictor omega-hydroxylation product of arachidonic acid that is produced by cytochrome P450 (CYP) enzymes, mainly by CYP4A and CYP4F isoforms. Arachidonic Acid 94-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 23013331-6 2012 20-Hydroxyeicosatetraenoic acid (20-HETE) is a vasoconstrictor omega-hydroxylation product of arachidonic acid that is produced by cytochrome P450 (CYP) enzymes, mainly by CYP4A and CYP4F isoforms. Arachidonic Acid 94-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 22087535-1 2012 Compounds that coordinate to the heme-iron of cytochrome P450 (CYP) enzymes are assumed to increase metabolic stability. Heme 33-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 22087535-1 2012 Compounds that coordinate to the heme-iron of cytochrome P450 (CYP) enzymes are assumed to increase metabolic stability. Heme 33-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 22087535-1 2012 Compounds that coordinate to the heme-iron of cytochrome P450 (CYP) enzymes are assumed to increase metabolic stability. Iron 38-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 22087535-1 2012 Compounds that coordinate to the heme-iron of cytochrome P450 (CYP) enzymes are assumed to increase metabolic stability. Iron 38-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 22687469-1 2012 Mammalian cytochrome P450 (CYP) comprise a large group of enzymes that play many important roles in the biosynthesis of steroid hormones and vitamins. Steroids 120-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 22687469-4 2012 In recent years, however, significant progress has been made in the X-ray crystallographic analysis of mammalian CYP enzymes involved in the steroid hormone and vitamin D(3) metabolism. Steroids 141-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 22687469-4 2012 In recent years, however, significant progress has been made in the X-ray crystallographic analysis of mammalian CYP enzymes involved in the steroid hormone and vitamin D(3) metabolism. Vitamin D 161-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 21594721-1 2012 PURPOSE: To assess the inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin (17-AG) on cytochrome P450 (CYP) enzymes. tanespimycin 61-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 21594721-1 2012 PURPOSE: To assess the inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin (17-AG) on cytochrome P450 (CYP) enzymes. tanespimycin 61-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 21594721-1 2012 PURPOSE: To assess the inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin (17-AG) on cytochrome P450 (CYP) enzymes. 17-Amino-17-demethoxygeldanamycin 100-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 21594721-1 2012 PURPOSE: To assess the inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin (17-AG) on cytochrome P450 (CYP) enzymes. 17-Amino-17-demethoxygeldanamycin 100-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 21594721-1 2012 PURPOSE: To assess the inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin (17-AG) on cytochrome P450 (CYP) enzymes. 17-AG 135-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 21594721-1 2012 PURPOSE: To assess the inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin (17-AG) on cytochrome P450 (CYP) enzymes. 17-AG 135-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 21594721-2 2012 METHODS: The inhibitory effect of tanespimycin and 17-AG on various CYP enzymes was determined in human liver microsomes. tanespimycin 34-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 21594721-2 2012 METHODS: The inhibitory effect of tanespimycin and 17-AG on various CYP enzymes was determined in human liver microsomes. 17-AG 51-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 21626050-0 2012 Docetaxel pharmacokinetics and its correlation with two in vivo probes for cytochrome P450 enzymes: the C(14)-erythromycin breath test and the antipyrine clearance test. Docetaxel 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 23018445-3 2012 In this study, the metabolic activities of three major hepatic CYP isoforms (2C19, 2D6, and 3A4) were investigated on structurally different central nervous system (CNS) acting drugs, amitriptyline, fluphenazine, and dothiepin. Amitriptyline 184-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 23018445-3 2012 In this study, the metabolic activities of three major hepatic CYP isoforms (2C19, 2D6, and 3A4) were investigated on structurally different central nervous system (CNS) acting drugs, amitriptyline, fluphenazine, and dothiepin. Fluphenazine 199-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 23018445-3 2012 In this study, the metabolic activities of three major hepatic CYP isoforms (2C19, 2D6, and 3A4) were investigated on structurally different central nervous system (CNS) acting drugs, amitriptyline, fluphenazine, and dothiepin. Dothiepin 217-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 22975950-0 2012 Effect of cytochrome P450-dependent epoxyeicosanoids on Ristocetin-induced thrombocyte aggregation. epoxyeicosanoids 36-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 22701192-7 2012 Concomitant administration of medications such as triazole antifungals which influence the cytochrome P-450 system can exacerbate this potential complication of ATRA. Triazoles 50-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 22701192-7 2012 Concomitant administration of medications such as triazole antifungals which influence the cytochrome P-450 system can exacerbate this potential complication of ATRA. Tretinoin 161-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 22856659-5 2012 Cytochrome P450 (CYP450)-mediated cocaine metabolism yields the reactive pro-oxidant compound norcocaine (NCOC) and further oxidative metabolites. Cocaine 34-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22856659-5 2012 Cytochrome P450 (CYP450)-mediated cocaine metabolism yields the reactive pro-oxidant compound norcocaine (NCOC) and further oxidative metabolites. Cocaine 34-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 22856659-5 2012 Cytochrome P450 (CYP450)-mediated cocaine metabolism yields the reactive pro-oxidant compound norcocaine (NCOC) and further oxidative metabolites. norcocaine 94-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 22856659-5 2012 Cytochrome P450 (CYP450)-mediated cocaine metabolism yields the reactive pro-oxidant compound norcocaine (NCOC) and further oxidative metabolites. norcocaine 94-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-23 22975950-0 2012 Effect of cytochrome P450-dependent epoxyeicosanoids on Ristocetin-induced thrombocyte aggregation. Ristocetin 56-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 22975950-1 2012 Epoxyeicosatrienoic acids (EETs) produced by cytochrome P450 (CYP)-dependent epoxidation of arachidonic acid (AA) inhibit thrombocyte adhesion to the vascular wall. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 22975950-1 2012 Epoxyeicosatrienoic acids (EETs) produced by cytochrome P450 (CYP)-dependent epoxidation of arachidonic acid (AA) inhibit thrombocyte adhesion to the vascular wall. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 22975950-1 2012 Epoxyeicosatrienoic acids (EETs) produced by cytochrome P450 (CYP)-dependent epoxidation of arachidonic acid (AA) inhibit thrombocyte adhesion to the vascular wall. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 22975950-1 2012 Epoxyeicosatrienoic acids (EETs) produced by cytochrome P450 (CYP)-dependent epoxidation of arachidonic acid (AA) inhibit thrombocyte adhesion to the vascular wall. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 21989950-4 2012 A series of in vitro studies were conducted to identify the human esterases involved in the prodrug hydrolysis and to identify the primary human cytochrome P450 and sulfotransferase (SULT) enzymes involved in the metabolism of brivanib. brivanib 227-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-181 22975950-1 2012 Epoxyeicosatrienoic acids (EETs) produced by cytochrome P450 (CYP)-dependent epoxidation of arachidonic acid (AA) inhibit thrombocyte adhesion to the vascular wall. Arachidonic Acid 92-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 22041137-2 2012 Tamoxifen requires metabolic activation by cytochrome P450 (CYP) enzymes for formation of active metabolites, 4-hydroxytamoxifen and endoxifen, which have 30- to 100-fold greater affinity to the estrogen receptor and the potency to suppress estrogen-dependent breast cancer cell proliferation. Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 22975950-1 2012 Epoxyeicosatrienoic acids (EETs) produced by cytochrome P450 (CYP)-dependent epoxidation of arachidonic acid (AA) inhibit thrombocyte adhesion to the vascular wall. Arachidonic Acid 92-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 22041137-2 2012 Tamoxifen requires metabolic activation by cytochrome P450 (CYP) enzymes for formation of active metabolites, 4-hydroxytamoxifen and endoxifen, which have 30- to 100-fold greater affinity to the estrogen receptor and the potency to suppress estrogen-dependent breast cancer cell proliferation. Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 22123124-1 2012 Cytochrome P450 oxidoreductase (POR) transfers electrons from NADPH to several oxygenase enzymes including cytochrome P450 (CYP). NADP 62-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 22041137-2 2012 Tamoxifen requires metabolic activation by cytochrome P450 (CYP) enzymes for formation of active metabolites, 4-hydroxytamoxifen and endoxifen, which have 30- to 100-fold greater affinity to the estrogen receptor and the potency to suppress estrogen-dependent breast cancer cell proliferation. hydroxytamoxifen 110-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 22975950-11 2012 These results indicate a highly specific role of CYP-eicosanoids in preventing thromboembolic events and suggest that the formation of 17,18-EEQ and 19,20-EDP may contribute to the anti-thrombotic effects of omega-3 fatty acids. Eicosanoids 53-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 22041137-2 2012 Tamoxifen requires metabolic activation by cytochrome P450 (CYP) enzymes for formation of active metabolites, 4-hydroxytamoxifen and endoxifen, which have 30- to 100-fold greater affinity to the estrogen receptor and the potency to suppress estrogen-dependent breast cancer cell proliferation. hydroxytamoxifen 110-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 22041137-2 2012 Tamoxifen requires metabolic activation by cytochrome P450 (CYP) enzymes for formation of active metabolites, 4-hydroxytamoxifen and endoxifen, which have 30- to 100-fold greater affinity to the estrogen receptor and the potency to suppress estrogen-dependent breast cancer cell proliferation. 4-hydroxy-N-desmethyltamoxifen 133-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 22041137-2 2012 Tamoxifen requires metabolic activation by cytochrome P450 (CYP) enzymes for formation of active metabolites, 4-hydroxytamoxifen and endoxifen, which have 30- to 100-fold greater affinity to the estrogen receptor and the potency to suppress estrogen-dependent breast cancer cell proliferation. 4-hydroxy-N-desmethyltamoxifen 133-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 22123124-1 2012 Cytochrome P450 oxidoreductase (POR) transfers electrons from NADPH to several oxygenase enzymes including cytochrome P450 (CYP). NADP 62-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 22041137-4 2012 The genetic polymorphisms in the other drug-metabolizing enzymes, including other CYP isoforms, sulfotransferases and UDP-glucuronosyltransferases might contribute to individual differences in the tamoxifen metabolism and clinical outcome of tamoxifen therapy although their contributions would be small. Tamoxifen 197-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 22975950-11 2012 These results indicate a highly specific role of CYP-eicosanoids in preventing thromboembolic events and suggest that the formation of 17,18-EEQ and 19,20-EDP may contribute to the anti-thrombotic effects of omega-3 fatty acids. Fatty Acids, Omega-3 208-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 22041137-4 2012 The genetic polymorphisms in the other drug-metabolizing enzymes, including other CYP isoforms, sulfotransferases and UDP-glucuronosyltransferases might contribute to individual differences in the tamoxifen metabolism and clinical outcome of tamoxifen therapy although their contributions would be small. Tamoxifen 242-251 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 22903709-1 2012 In this chapter we explore the inducible cytochrome P450 (CYP) forms as an example of membrane proteins analysis that relies on sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) fractionation with subsequent mass spectrometric (MS) identification. Sodium Dodecyl Sulfate 128-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 22100140-1 2012 Because of the increasing number of immunocompromised patients and due to problems with antifungal treatment, especially with the most widely used antifungals, azoles, there is an urgent need for new, potent and safe antifungals with fewer cytochrome P450 (CYP)-mediated interactions with other drugs. Azoles 160-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-255 22100140-1 2012 Because of the increasing number of immunocompromised patients and due to problems with antifungal treatment, especially with the most widely used antifungals, azoles, there is an urgent need for new, potent and safe antifungals with fewer cytochrome P450 (CYP)-mediated interactions with other drugs. Azoles 160-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 257-260 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). Arachidonic Acid 136-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). Arachidonic Acid 136-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). dihydroxyeicosatrienoic acids 256-285 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). dihydroxyeicosatrienoic acids 256-285 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). dhets 287-292 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 22848834-1 2012 Epoxyeicosatrienoic acids (EETs) are generated by the activity of both selective and also more general cytochrome p450 (CYP) enzymes on arachidonic acid and inactivated largely by soluble epoxide hydrolase (sEH), which converts them to their corresponding dihydroxyeicosatrienoic acids (DHETs). dhets 287-292 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 22182836-1 2012 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases have beneficial effects in certain cardiovascular and kidney diseases. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 22182836-1 2012 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases have beneficial effects in certain cardiovascular and kidney diseases. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 22182836-1 2012 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases have beneficial effects in certain cardiovascular and kidney diseases. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 22182836-1 2012 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases have beneficial effects in certain cardiovascular and kidney diseases. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 22182836-1 2012 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases have beneficial effects in certain cardiovascular and kidney diseases. Arachidonic Acid 48-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 22182836-1 2012 Epoxyeicosatrienoic acids (EETs) generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases have beneficial effects in certain cardiovascular and kidney diseases. Arachidonic Acid 48-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 22903709-1 2012 In this chapter we explore the inducible cytochrome P450 (CYP) forms as an example of membrane proteins analysis that relies on sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) fractionation with subsequent mass spectrometric (MS) identification. Sodium Dodecyl Sulfate 128-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 22903709-1 2012 In this chapter we explore the inducible cytochrome P450 (CYP) forms as an example of membrane proteins analysis that relies on sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) fractionation with subsequent mass spectrometric (MS) identification. polyacrylamide 151-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 22903709-1 2012 In this chapter we explore the inducible cytochrome P450 (CYP) forms as an example of membrane proteins analysis that relies on sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) fractionation with subsequent mass spectrometric (MS) identification. polyacrylamide 151-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 22903709-1 2012 In this chapter we explore the inducible cytochrome P450 (CYP) forms as an example of membrane proteins analysis that relies on sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) fractionation with subsequent mass spectrometric (MS) identification. Sodium Dodecyl Sulfate 187-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 22903709-1 2012 In this chapter we explore the inducible cytochrome P450 (CYP) forms as an example of membrane proteins analysis that relies on sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) fractionation with subsequent mass spectrometric (MS) identification. Sodium Dodecyl Sulfate 187-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 22778656-3 2012 A model used to effectively describe the cytochrome P450 deposition onto carbon nanotubes was confirmed by Monte Carlo simulations. Carbon 73-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 25298920-2 2012 In the last decade there has been an increase in realization of the power of CYP biocatalysts for detoxification of soil and water contaminants using transgenic plants. Water 125-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 22079551-1 2012 Cytochrome P450 family 4 (CYP4) proteins metabolize fatty acids, eicosanoids, and vitamin D and are important for chemical defense. Fatty Acids 52-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-24 22079551-1 2012 Cytochrome P450 family 4 (CYP4) proteins metabolize fatty acids, eicosanoids, and vitamin D and are important for chemical defense. Fatty Acids 52-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-30 22079551-1 2012 Cytochrome P450 family 4 (CYP4) proteins metabolize fatty acids, eicosanoids, and vitamin D and are important for chemical defense. Eicosanoids 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-24 22079551-1 2012 Cytochrome P450 family 4 (CYP4) proteins metabolize fatty acids, eicosanoids, and vitamin D and are important for chemical defense. Eicosanoids 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-30 22079551-1 2012 Cytochrome P450 family 4 (CYP4) proteins metabolize fatty acids, eicosanoids, and vitamin D and are important for chemical defense. Vitamin D 82-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-24 22079551-1 2012 Cytochrome P450 family 4 (CYP4) proteins metabolize fatty acids, eicosanoids, and vitamin D and are important for chemical defense. Vitamin D 82-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-30 22778656-0 2012 Electrochemical detection of anti-breast-cancer agents in human serum by cytochrome P450-coated carbon nanotubes. Carbon 96-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 21880455-0 2011 Rapid simultaneous analysis of cyclooxygenase, lipoxygenase and cytochrome P-450 metabolites of arachidonic and linoleic acids using high performance liquid chromatography/mass spectrometry in tandem mode. arachidonic and linoleic acids 96-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 22778656-2 2012 Multi-walled carbon nanotubes were functionalized with three different cytochrome P450 isoforms (CYP1A2, CYP2B6, and CYP3A4). Carbon 13-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 21946300-0 2011 Human cytochrome-P450 enzymes metabolize N-(2-methoxyphenyl)hydroxylamine, a metabolite of the carcinogens o-anisidine and o-nitroanisole, thereby dictating its genotoxicity. N-(2-methoxyphenyl)hydroxylamine 41-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 21946300-0 2011 Human cytochrome-P450 enzymes metabolize N-(2-methoxyphenyl)hydroxylamine, a metabolite of the carcinogens o-anisidine and o-nitroanisole, thereby dictating its genotoxicity. 2-anisidine 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 21946300-0 2011 Human cytochrome-P450 enzymes metabolize N-(2-methoxyphenyl)hydroxylamine, a metabolite of the carcinogens o-anisidine and o-nitroanisole, thereby dictating its genotoxicity. 2-nitroanisole 123-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 21880455-1 2011 Eicosanoids are oxidized arachidonate-derived lipid products generated by cyclooxygenase, lipoxygenase and cytochrome P-450 pathways. Eicosanoids 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-123 21880455-1 2011 Eicosanoids are oxidized arachidonate-derived lipid products generated by cyclooxygenase, lipoxygenase and cytochrome P-450 pathways. Arachidonic Acid 25-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-123 21411308-1 2011 Protein engineering of cytochrome P450 monooxygenases (P450s) has been very successful in generating valuable non-natural activities and properties, allowing these powerful catalysts to be used for the synthesis of drug metabolites and in biosynthetic pathways for the production of precursors of artemisinin and paclitaxel. artemisinin 297-308 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 21689071-0 2011 Cytochrome P450 metabolites of arachidonic acid are elevated in stroke patients compared with healthy controls. Arachidonic Acid 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21689071-1 2011 CYP450AAM [arachidonic acid metabolites of the CYP450 (cytochrome P450) enzyme system] have a range of biological functions. Arachidonic Acid 11-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 21689071-1 2011 CYP450AAM [arachidonic acid metabolites of the CYP450 (cytochrome P450) enzyme system] have a range of biological functions. Arachidonic Acid 11-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 22048864-2 2011 Changes in dietary fatty acids, specifically the polyunsaturated fatty acids of the omega-3 and omega-6 families and some derived eicosanoids from lipoxygenases, cyclooxygenases, and cytochrome P-450, seem to control the activity of transcription factor families involved in cancer cell proliferation or cell death. Fatty Acids 19-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 22048864-2 2011 Changes in dietary fatty acids, specifically the polyunsaturated fatty acids of the omega-3 and omega-6 families and some derived eicosanoids from lipoxygenases, cyclooxygenases, and cytochrome P-450, seem to control the activity of transcription factor families involved in cancer cell proliferation or cell death. Fatty Acids, Unsaturated 49-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 22048864-2 2011 Changes in dietary fatty acids, specifically the polyunsaturated fatty acids of the omega-3 and omega-6 families and some derived eicosanoids from lipoxygenases, cyclooxygenases, and cytochrome P-450, seem to control the activity of transcription factor families involved in cancer cell proliferation or cell death. Eicosanoids 130-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 21411308-1 2011 Protein engineering of cytochrome P450 monooxygenases (P450s) has been very successful in generating valuable non-natural activities and properties, allowing these powerful catalysts to be used for the synthesis of drug metabolites and in biosynthetic pathways for the production of precursors of artemisinin and paclitaxel. Paclitaxel 313-323 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 21995615-0 2011 Triazole antifungal agents drug-drug interactions involving hepatic cytochrome P450. Triazoles 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 22089302-11 2011 Results suggest the CYP metabolites (i.e., DHETs) are released during short-term high-intensity and long-term moderate-intensity exercise. dhets 43-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-23 21964476-4 2011 DCPT toxicity is dependent upon the presence of an intact TZD ring and cytochrome P450 (CYP)-mediated biotransformation. 3-(3,5-dichlorophenyl)-2,4-thiazolidinedione 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 21964476-4 2011 DCPT toxicity is dependent upon the presence of an intact TZD ring and cytochrome P450 (CYP)-mediated biotransformation. 3-(3,5-dichlorophenyl)-2,4-thiazolidinedione 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 21901220-0 2011 Oxidation of 10-undecenoic acid by cytochrome P450(BM-3) and its Compound I transient. undecylenic acid 13-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-55 21901220-1 2011 Oxidations of 10-undecenoic acid by cytochrome P450(BM-3) and its Compound I transient were studied. undecylenic acid 14-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-56 21350850-1 2011 BACKGROUND: In vitro data indicate that the sorafenib is a moderate inhibitor of cytochrome P450 (CYP) enzymes, including CYP3A4, CYP2C19, and CYP2D6. Sorafenib 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 21350850-1 2011 BACKGROUND: In vitro data indicate that the sorafenib is a moderate inhibitor of cytochrome P450 (CYP) enzymes, including CYP3A4, CYP2C19, and CYP2D6. Sorafenib 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 21995615-2 2011 As substrates and inhibitors of cytochrome P450 (CYP), the triazoles can interact with many drugs, which may lead to enhanced toxicity of the concomitant medication(s) or ineffective antifungal treatment. Triazoles 59-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 21995615-2 2011 As substrates and inhibitors of cytochrome P450 (CYP), the triazoles can interact with many drugs, which may lead to enhanced toxicity of the concomitant medication(s) or ineffective antifungal treatment. Triazoles 59-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 21995615-4 2011 AREAS COVERED: This manuscript reviews the role of human hepatic CYP in triazole-drug interactions, illustrates how recent discoveries in pharmacogenetics and triazole metabolism impact our understanding of these interactions, and summarizes the most clinically important triazole-drug interactions. Triazoles 72-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 21995615-4 2011 AREAS COVERED: This manuscript reviews the role of human hepatic CYP in triazole-drug interactions, illustrates how recent discoveries in pharmacogenetics and triazole metabolism impact our understanding of these interactions, and summarizes the most clinically important triazole-drug interactions. Triazoles 159-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 21995615-4 2011 AREAS COVERED: This manuscript reviews the role of human hepatic CYP in triazole-drug interactions, illustrates how recent discoveries in pharmacogenetics and triazole metabolism impact our understanding of these interactions, and summarizes the most clinically important triazole-drug interactions. Triazoles 159-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 21995615-5 2011 A search of English language, original research and scholarly reviews describing interactions between triazole antifungal agents and human CYP published from 1980 to present was undertaken using PubMed. Triazoles 102-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 21995615-6 2011 EXPERT OPINION: Triazoles interact with many drugs and the primary mechanism for these interactions is hepatic CYP. Triazoles 16-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-114 21995615-8 2011 However, advances in genotyping, improved analytical technology and bioanalytical methods, which enable accurate molecular identification and stereochemical analysis, have substantially added to the understanding of the role hepatic CYP plays in triazole-drug interactions in humans. Triazoles 246-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 233-236 21996372-2 2011 TRPV4 is also thermosensitive and responds to moderate heat (from 24 to 27 C) as well as to phorbol esters (4alpha-PDD) and several endogenous substances including arachidonic acid (AA), the endocannabinoids anandamide and 2-AG, and cytochrome P-450 metabolites of AA, such as epoxyeicosatrienoic acids. Phorbol Esters 93-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-250 26598170-0 2011 Do Two Different Reaction Mechanisms Contribute to the Hydroxylation of Primary Amines by Cytochrome P450? Amines 80-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 21864702-3 2011 A well-studied group of autacoid mediators that are the products of arachidonic acid metabolism include: the prostaglandins, leukotrienes, lipoxins and cytochrome P450 (CYP) derived bioactive products. Arachidonic Acid 68-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-167 21864702-3 2011 A well-studied group of autacoid mediators that are the products of arachidonic acid metabolism include: the prostaglandins, leukotrienes, lipoxins and cytochrome P450 (CYP) derived bioactive products. Arachidonic Acid 68-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 27147854-0 2011 Does cytochrome P450 liver isoenzyme induction increase the risk of liver toxicity after paracetamol overdose? Acetaminophen 89-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-20 27147854-3 2011 These include the dose of paracetamol ingested, time to presentation, decreased liver glutathione, and induction of cytochrome P450 (CYP) isoenzymes responsible for the metabolism of paracetamol to its toxic metabolite N-acetyl-p-benzoquinoneimine (NAPQI). Acetaminophen 183-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 27147854-3 2011 These include the dose of paracetamol ingested, time to presentation, decreased liver glutathione, and induction of cytochrome P450 (CYP) isoenzymes responsible for the metabolism of paracetamol to its toxic metabolite N-acetyl-p-benzoquinoneimine (NAPQI). Acetaminophen 183-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 27147854-3 2011 These include the dose of paracetamol ingested, time to presentation, decreased liver glutathione, and induction of cytochrome P450 (CYP) isoenzymes responsible for the metabolism of paracetamol to its toxic metabolite N-acetyl-p-benzoquinoneimine (NAPQI). N-acetyl-4-benzoquinoneimine 219-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 27147854-3 2011 These include the dose of paracetamol ingested, time to presentation, decreased liver glutathione, and induction of cytochrome P450 (CYP) isoenzymes responsible for the metabolism of paracetamol to its toxic metabolite N-acetyl-p-benzoquinoneimine (NAPQI). N-acetyl-4-benzoquinoneimine 219-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 27147854-3 2011 These include the dose of paracetamol ingested, time to presentation, decreased liver glutathione, and induction of cytochrome P450 (CYP) isoenzymes responsible for the metabolism of paracetamol to its toxic metabolite N-acetyl-p-benzoquinoneimine (NAPQI). N-acetyl-4-benzoquinoneimine 249-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 27147854-3 2011 These include the dose of paracetamol ingested, time to presentation, decreased liver glutathione, and induction of cytochrome P450 (CYP) isoenzymes responsible for the metabolism of paracetamol to its toxic metabolite N-acetyl-p-benzoquinoneimine (NAPQI). N-acetyl-4-benzoquinoneimine 249-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 27147854-4 2011 In this paper, we review the currently published literature to determine whether induction of relevant CYP isoenzymes is a risk factor for hepatotoxicity in patients with acute paracetamol overdose. Acetaminophen 177-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 26598170-1 2011 Three possible mechanisms have been suggested for the hydroxylation of primary and secondary amines by the cytochrome P450 enzyme family. Amines 93-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 27147854-5 2011 Animal and human in vitro studies have shown that the CYP isoenzyme responsible for the majority of human biotransformation of paracetamol to NAPQI is CYP2E1 at both therapeutic and toxic doses of paracetamol. Acetaminophen 127-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 21970838-1 2011 AIM: To investigate the metabolism of 3-cyanomethyl-4-methyl-DCK (CMDCK), a novel anti-HIV agent, by human liver microsomes (HLMs) and recombinant cytochrome P450 enzymes (CYPs). 3-cyanomethyl 38-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-162 27147854-5 2011 Animal and human in vitro studies have shown that the CYP isoenzyme responsible for the majority of human biotransformation of paracetamol to NAPQI is CYP2E1 at both therapeutic and toxic doses of paracetamol. N-acetyl-4-benzoquinoneimine 142-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 27147854-5 2011 Animal and human in vitro studies have shown that the CYP isoenzyme responsible for the majority of human biotransformation of paracetamol to NAPQI is CYP2E1 at both therapeutic and toxic doses of paracetamol. Acetaminophen 197-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 21854846-0 2011 Risk assessment of accidental nortriptyline poisoning: the importance of cytochrome P450 for nortriptyline elimination investigated using a population-based pharmacokinetic simulator. Nortriptyline 93-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 21854846-1 2011 It is not possible to make a prospective clinical study that reveals the importance of the nortriptyline metabolising cytochrome P450 (CYP) isoforms (CYP1A2, CYP2C19, CYP2D6, and CYP3A4) in relation to attaining potential toxic nortriptyline concentrations with a possibly fatal outcome. Nortriptyline 91-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 21854846-1 2011 It is not possible to make a prospective clinical study that reveals the importance of the nortriptyline metabolising cytochrome P450 (CYP) isoforms (CYP1A2, CYP2C19, CYP2D6, and CYP3A4) in relation to attaining potential toxic nortriptyline concentrations with a possibly fatal outcome. Nortriptyline 91-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 21854846-1 2011 It is not possible to make a prospective clinical study that reveals the importance of the nortriptyline metabolising cytochrome P450 (CYP) isoforms (CYP1A2, CYP2C19, CYP2D6, and CYP3A4) in relation to attaining potential toxic nortriptyline concentrations with a possibly fatal outcome. Nortriptyline 228-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. 8-hydroxyloxapine 142-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. 7-hydroxyloxapine 161-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 21826677-0 2011 In vitro identification of the human cytochrome p450 enzymes involved in the oxidative metabolism of loxapine. Loxapine 101-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. 7-hydroxyloxapine 161-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. Loxapine 130-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. n-desmethylloxapine 180-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. Loxapine 130-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. n-desmethylloxapine 180-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. 8-hydroxyloxapine 142-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 21352268-2 2011 The main enzyme responsible for activating clopidogrel is the cytochrome P450 (CYP) isoenzyme CYP2C19, which is polymorphic. Clopidogrel 43-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 21352268-2 2011 The main enzyme responsible for activating clopidogrel is the cytochrome P450 (CYP) isoenzyme CYP2C19, which is polymorphic. Clopidogrel 43-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 21352268-5 2011 In particular, omeprazole has been shown to inhibit the CYP-mediated metabolism of clopidogrel, and some studies have shown that the combination was associated with a higher incidence of cardiovascular adverse reactions than clopidogrel given alone. Omeprazole 15-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. Amoxapine 201-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 21352268-5 2011 In particular, omeprazole has been shown to inhibit the CYP-mediated metabolism of clopidogrel, and some studies have shown that the combination was associated with a higher incidence of cardiovascular adverse reactions than clopidogrel given alone. Clopidogrel 83-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. Amoxapine 201-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. Loxapine N-oxide 216-232 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 21826677-1 2011 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) enzymes responsible for the oxidative metabolism of loxapine to 8-hydroxyloxapine, 7-hydroxyloxapine, N-desmethylloxapine (amoxapine) and loxapine N-oxide. Loxapine N-oxide 216-232 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 21685474-2 2011 Previous studies showed that the presence of one or two minor alleles of the cytochrome P450, subfamily 2C polypeptide 8 gene (CYP2C8) polymorphism rs1934951 was an independent prognostic marker associated with development of osteonecrosis of the jaw in multiple myeloma patients treated with bisphosphonates. Diphosphonates 293-308 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-120 21826477-2 2011 The available thienopyridines are prodrugs and must be converted into active forms by the cytochrome P450 (CYP) enzyme system. Thienopyridines 14-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 21826477-2 2011 The available thienopyridines are prodrugs and must be converted into active forms by the cytochrome P450 (CYP) enzyme system. Thienopyridines 14-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-110 21826477-7 2011 Conversely, agents that induce CYP activity increase clopidogrel responsiveness. Clopidogrel 53-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 22009620-10 2011 The primary number of clinically relevant pharmacological interactions is correlated with modifications of biotransformation of drugs due to Cytochrome P450 (CYP) hepatic enzymes which are involved in oxidative drug processes, including lipophilic antimicrobial drugs such as the macrolides, the fluoroquinolones (to be considered amphoteric) and the antifungal azole derivatives. Macrolides 280-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-156 22009620-10 2011 The primary number of clinically relevant pharmacological interactions is correlated with modifications of biotransformation of drugs due to Cytochrome P450 (CYP) hepatic enzymes which are involved in oxidative drug processes, including lipophilic antimicrobial drugs such as the macrolides, the fluoroquinolones (to be considered amphoteric) and the antifungal azole derivatives. Macrolides 280-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-161 22009620-10 2011 The primary number of clinically relevant pharmacological interactions is correlated with modifications of biotransformation of drugs due to Cytochrome P450 (CYP) hepatic enzymes which are involved in oxidative drug processes, including lipophilic antimicrobial drugs such as the macrolides, the fluoroquinolones (to be considered amphoteric) and the antifungal azole derivatives. Fluoroquinolones 296-312 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-156 22009620-10 2011 The primary number of clinically relevant pharmacological interactions is correlated with modifications of biotransformation of drugs due to Cytochrome P450 (CYP) hepatic enzymes which are involved in oxidative drug processes, including lipophilic antimicrobial drugs such as the macrolides, the fluoroquinolones (to be considered amphoteric) and the antifungal azole derivatives. Fluoroquinolones 296-312 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-161 22009620-10 2011 The primary number of clinically relevant pharmacological interactions is correlated with modifications of biotransformation of drugs due to Cytochrome P450 (CYP) hepatic enzymes which are involved in oxidative drug processes, including lipophilic antimicrobial drugs such as the macrolides, the fluoroquinolones (to be considered amphoteric) and the antifungal azole derivatives. Azoles 362-367 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-156 22009620-10 2011 The primary number of clinically relevant pharmacological interactions is correlated with modifications of biotransformation of drugs due to Cytochrome P450 (CYP) hepatic enzymes which are involved in oxidative drug processes, including lipophilic antimicrobial drugs such as the macrolides, the fluoroquinolones (to be considered amphoteric) and the antifungal azole derivatives. Azoles 362-367 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-161 21564387-1 2011 BACKGROUND: Phenobarbital induces specific hepatic cytochrome P-450 enzyme pathways causing increased clearance of hepatically metabolized drugs. Phenobarbital 12-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Codeine 93-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 22145115-0 2011 Study on cytochrome p-450 dependent retinoic Acid metabolism and its inhibitors as potential agents for cancer therapy. Tretinoin 36-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 22145115-13 2011 1-Substituted imidazoles bind to cytochrome P-450 with a very high affinity but substitution in the other position of the imidazole decreases the binding affinity. 1-substituted imidazoles 0-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 22145115-13 2011 1-Substituted imidazoles bind to cytochrome P-450 with a very high affinity but substitution in the other position of the imidazole decreases the binding affinity. imidazole 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 21726172-2 2011 Due to the potential for drug-drug interactions, the ability of toremifene and its primary circulating metabolite N-desmethyltoremifene (NDMT) to inhibit nine human cytochrome P450 (CYP) enzymes was determined using human liver microsomes. Toremifene 64-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-185 21726172-2 2011 Due to the potential for drug-drug interactions, the ability of toremifene and its primary circulating metabolite N-desmethyltoremifene (NDMT) to inhibit nine human cytochrome P450 (CYP) enzymes was determined using human liver microsomes. N-desmethyltoremifene 114-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-180 21726172-2 2011 Due to the potential for drug-drug interactions, the ability of toremifene and its primary circulating metabolite N-desmethyltoremifene (NDMT) to inhibit nine human cytochrome P450 (CYP) enzymes was determined using human liver microsomes. N-desmethyltoremifene 114-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-185 21726172-2 2011 Due to the potential for drug-drug interactions, the ability of toremifene and its primary circulating metabolite N-desmethyltoremifene (NDMT) to inhibit nine human cytochrome P450 (CYP) enzymes was determined using human liver microsomes. N-desmethyltoremifene 137-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-180 21726172-2 2011 Due to the potential for drug-drug interactions, the ability of toremifene and its primary circulating metabolite N-desmethyltoremifene (NDMT) to inhibit nine human cytochrome P450 (CYP) enzymes was determined using human liver microsomes. N-desmethyltoremifene 137-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-185 21726172-6 2011 CYP inhibition by NDMT was similar in magnitude to toremifene. N-desmethyltoremifene 18-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21496210-2 2011 In this study, the interaction of degarelix with human cytochrome P450 (CYP450) enzymes was investigated in vitro. acetyl-2-naphthylalanyl-3-chlorophenylalanyl-1-oxohexadecyl-seryl-4-aminophenylalanyl(hydroorotyl)-4-aminophenylalanyl(carbamoyl)-leucyl-ILys-prolyl-alaninamide 34-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 21496210-2 2011 In this study, the interaction of degarelix with human cytochrome P450 (CYP450) enzymes was investigated in vitro. acetyl-2-naphthylalanyl-3-chlorophenylalanyl-1-oxohexadecyl-seryl-4-aminophenylalanyl(hydroorotyl)-4-aminophenylalanyl(carbamoyl)-leucyl-ILys-prolyl-alaninamide 34-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-78 21496210-6 2011 Induction of CYP450 enzyme activity by degarelix was investigated using primary human hepatocytes. acetyl-2-naphthylalanyl-3-chlorophenylalanyl-1-oxohexadecyl-seryl-4-aminophenylalanyl(hydroorotyl)-4-aminophenylalanyl(carbamoyl)-leucyl-ILys-prolyl-alaninamide 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-19 21496210-10 2011 Degarelix appears to be a poor substrate of the CYP450 enzyme system, and the in vitro results indicate that the interaction between CYP450 and degarelix is low. acetyl-2-naphthylalanyl-3-chlorophenylalanyl-1-oxohexadecyl-seryl-4-aminophenylalanyl(hydroorotyl)-4-aminophenylalanyl(carbamoyl)-leucyl-ILys-prolyl-alaninamide 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-54 21496210-10 2011 Degarelix appears to be a poor substrate of the CYP450 enzyme system, and the in vitro results indicate that the interaction between CYP450 and degarelix is low. acetyl-2-naphthylalanyl-3-chlorophenylalanyl-1-oxohexadecyl-seryl-4-aminophenylalanyl(hydroorotyl)-4-aminophenylalanyl(carbamoyl)-leucyl-ILys-prolyl-alaninamide 144-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-54 21496210-10 2011 Degarelix appears to be a poor substrate of the CYP450 enzyme system, and the in vitro results indicate that the interaction between CYP450 and degarelix is low. acetyl-2-naphthylalanyl-3-chlorophenylalanyl-1-oxohexadecyl-seryl-4-aminophenylalanyl(hydroorotyl)-4-aminophenylalanyl(carbamoyl)-leucyl-ILys-prolyl-alaninamide 144-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-139 21726172-0 2011 Drug interaction potential of toremifene and N-desmethyltoremifene with multiple cytochrome P450 isoforms. Toremifene 30-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 21726172-0 2011 Drug interaction potential of toremifene and N-desmethyltoremifene with multiple cytochrome P450 isoforms. N-desmethyltoremifene 45-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 21726172-2 2011 Due to the potential for drug-drug interactions, the ability of toremifene and its primary circulating metabolite N-desmethyltoremifene (NDMT) to inhibit nine human cytochrome P450 (CYP) enzymes was determined using human liver microsomes. Toremifene 64-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-180 21870861-0 2011 A mechanistic hypothesis for the cytochrome P450-catalyzed cis-trans isomerization of 4-hydroxytamoxifen: an unusual redox reaction. hydroxytamoxifen 86-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 21870861-1 2011 We provide a detailed description of the cis-trans isomerization of 4-hydroxytamoxifen/endoxifen catalyzed by several isoforms from the cytochrome P450 (CYP) superfamily, including CYP1B1, CYP2B6, and CYP2C19. hydroxytamoxifen 68-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-151 21870861-1 2011 We provide a detailed description of the cis-trans isomerization of 4-hydroxytamoxifen/endoxifen catalyzed by several isoforms from the cytochrome P450 (CYP) superfamily, including CYP1B1, CYP2B6, and CYP2C19. hydroxytamoxifen 68-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-156 21870861-1 2011 We provide a detailed description of the cis-trans isomerization of 4-hydroxytamoxifen/endoxifen catalyzed by several isoforms from the cytochrome P450 (CYP) superfamily, including CYP1B1, CYP2B6, and CYP2C19. 4-hydroxy-N-desmethyltamoxifen 87-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-151 21870861-1 2011 We provide a detailed description of the cis-trans isomerization of 4-hydroxytamoxifen/endoxifen catalyzed by several isoforms from the cytochrome P450 (CYP) superfamily, including CYP1B1, CYP2B6, and CYP2C19. 4-hydroxy-N-desmethyltamoxifen 87-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-156 21798277-1 2011 The environmental pollutant 7H-dibenzo[c,g]carbazole (DBC) and its derivative, 5,9-dimethylDBC (DiMeDBC), produced significant and dose-dependent levels of micronuclei followed by a substantial increase in the frequency of apoptotic cells in the V79MZh3A4 cell line stably expressing the human cytochrome P450 (hCYP) 3A4. 7H-dibenzo(c,g)carbazole 28-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-320 21798277-1 2011 The environmental pollutant 7H-dibenzo[c,g]carbazole (DBC) and its derivative, 5,9-dimethylDBC (DiMeDBC), produced significant and dose-dependent levels of micronuclei followed by a substantial increase in the frequency of apoptotic cells in the V79MZh3A4 cell line stably expressing the human cytochrome P450 (hCYP) 3A4. 7H-dibenzo(c,g)carbazole 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-320 21798277-1 2011 The environmental pollutant 7H-dibenzo[c,g]carbazole (DBC) and its derivative, 5,9-dimethylDBC (DiMeDBC), produced significant and dose-dependent levels of micronuclei followed by a substantial increase in the frequency of apoptotic cells in the V79MZh3A4 cell line stably expressing the human cytochrome P450 (hCYP) 3A4. 5,9-dimethyldbc 79-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-320 21798277-1 2011 The environmental pollutant 7H-dibenzo[c,g]carbazole (DBC) and its derivative, 5,9-dimethylDBC (DiMeDBC), produced significant and dose-dependent levels of micronuclei followed by a substantial increase in the frequency of apoptotic cells in the V79MZh3A4 cell line stably expressing the human cytochrome P450 (hCYP) 3A4. dimedbc 96-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-320 21763268-0 2011 C-26 vs. C-27 hydroxylation of insect steroid hormones: regioselectivity of a microsomal cytochrome P450 from a hormone-resistant cell line. Carbon 0-1 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 21763268-0 2011 C-26 vs. C-27 hydroxylation of insect steroid hormones: regioselectivity of a microsomal cytochrome P450 from a hormone-resistant cell line. Steroids 38-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 21763268-3 2011 26-Hydroxylation of the insect steroid hormone 20-hydroxyecdysone by a microsomal cytochrome P450 was previously found to be responsible for hormonal resistance in a Chironomus cell line mainly producing the (25S)-epimer of 20,26-dihydroxyecdysone. Steroids 31-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 21763268-3 2011 26-Hydroxylation of the insect steroid hormone 20-hydroxyecdysone by a microsomal cytochrome P450 was previously found to be responsible for hormonal resistance in a Chironomus cell line mainly producing the (25S)-epimer of 20,26-dihydroxyecdysone. Ecdysterone 47-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 21763268-3 2011 26-Hydroxylation of the insect steroid hormone 20-hydroxyecdysone by a microsomal cytochrome P450 was previously found to be responsible for hormonal resistance in a Chironomus cell line mainly producing the (25S)-epimer of 20,26-dihydroxyecdysone. 20,26-dihydroxyecdysone 224-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 21750172-1 2011 BACKGROUND: Tamoxifen is oxidized by cytochrome-P450 enzymes (e.g., CYP2D6) to two active metabolites, which are eliminated via glucuronidation by UDP-glucuronosyl transferases (UGT). Tamoxifen 12-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Codeine 93-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-83 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Oxycodone 102-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Oxycodone 102-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-83 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Hydrocodone 113-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Hydrocodone 113-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-83 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Fentanyl 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Fentanyl 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-83 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Tramadol 136-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Tramadol 136-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-83 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Methadone 150-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 21999760-5 2011 Opioids metabolized by the drug metabolizing enzymes of the cytochrome P450 (CYP450) system (codeine, oxycodone, hydrocodone, fentanyl, tramadol, and methadone) are associated with numerous DDIs that can result in either a reduction in opioid effect or excess opioid effects. Methadone 150-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-83 21999760-6 2011 Conversely, opioids that are not metabolized by that system (morphine, oxymorphone, and hydromorphone) tend to be involved in fewer CYP450-associated pharmacokinetic DDIs. Morphine 61-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-138 21999760-6 2011 Conversely, opioids that are not metabolized by that system (morphine, oxymorphone, and hydromorphone) tend to be involved in fewer CYP450-associated pharmacokinetic DDIs. Oxymorphone 71-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-138 21999760-6 2011 Conversely, opioids that are not metabolized by that system (morphine, oxymorphone, and hydromorphone) tend to be involved in fewer CYP450-associated pharmacokinetic DDIs. Hydromorphone 88-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-138 21876481-0 2011 Evaluation of the effects of Mitragyna speciosa alkaloid extract on cytochrome P450 enzymes using a high throughput assay. Alkaloids 48-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 21740382-1 2011 The original map of mammalian cytochrome P450 (CYP450) residues involved in substrate recognition was prepared for the CYP2 family by Gotoh in 1992 by manual alignment of mammalian CYP450 residues with substrate recognition site (SRS) residues manually delimited from a bacterial cytochrome P450-substrate complex. gotoh 134-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 21740382-1 2011 The original map of mammalian cytochrome P450 (CYP450) residues involved in substrate recognition was prepared for the CYP2 family by Gotoh in 1992 by manual alignment of mammalian CYP450 residues with substrate recognition site (SRS) residues manually delimited from a bacterial cytochrome P450-substrate complex. gotoh 134-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-53 21740382-1 2011 The original map of mammalian cytochrome P450 (CYP450) residues involved in substrate recognition was prepared for the CYP2 family by Gotoh in 1992 by manual alignment of mammalian CYP450 residues with substrate recognition site (SRS) residues manually delimited from a bacterial cytochrome P450-substrate complex. gotoh 134-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-187 21740382-1 2011 The original map of mammalian cytochrome P450 (CYP450) residues involved in substrate recognition was prepared for the CYP2 family by Gotoh in 1992 by manual alignment of mammalian CYP450 residues with substrate recognition site (SRS) residues manually delimited from a bacterial cytochrome P450-substrate complex. gotoh 134-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 280-295 21861666-1 2011 AIMS: To investigate associations between novel human cytochrome P450 (CYP450) combinatory (multigene) and substrate-specific drug metabolism indices, and elements of metabolic syndrome, such as low density lipoprotein cholesterol (LDLc), high density lipoprotein cholesterol (HDLc), triglycerides and BMI, using physiogenomic analysis. Triglycerides 284-297 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 21861666-1 2011 AIMS: To investigate associations between novel human cytochrome P450 (CYP450) combinatory (multigene) and substrate-specific drug metabolism indices, and elements of metabolic syndrome, such as low density lipoprotein cholesterol (LDLc), high density lipoprotein cholesterol (HDLc), triglycerides and BMI, using physiogenomic analysis. Triglycerides 284-297 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-77 21682551-6 2011 Pitavastatin has a distinctive metabolic profile that means it is marginally metabolised by CYP enzymes, and is therefore expected to have a low risk of DDIs and related ADRs. pitavastatin 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 21704641-0 2011 Identification of cytochrome P450 enzymes responsible for metabolism of cannabidiol by human liver microsomes. Cannabidiol 72-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 21694616-3 2011 In our previous study, we found that the cytochrome P-450 (CYP) isozyme 2B6 preferentially metabolizes the S-methadone enantiomer. Methadone 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 21694616-3 2011 In our previous study, we found that the cytochrome P-450 (CYP) isozyme 2B6 preferentially metabolizes the S-methadone enantiomer. Methadone 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 21704641-3 2011 In this study, we examined in vitro metabolism of CBD with human liver microsomes (HLMs) to clarify cytochrome P450 (CYP) isoforms involved in the CBD oxidations. Cannabidiol 50-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 21704641-3 2011 In this study, we examined in vitro metabolism of CBD with human liver microsomes (HLMs) to clarify cytochrome P450 (CYP) isoforms involved in the CBD oxidations. Cannabidiol 50-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-120 21704641-7 2011 Seven of 14 recombinant human CYP enzymes examined (CYP1A1, CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4, and CYP3A5) were capable of metabolizing CBD. Cannabidiol 142-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 21704641-8 2011 The correlations between CYP isoform-specific activities and CBD oxidative activities in 16 individual HLMs indicated that 6beta-OH- and 4""-OH-CBDs were mainly formed by CYP3A4, which was supported by inhibition studies using ketoconazole and an anti-CYP3A4 antibody. 6beta-oh- and 4""-oh-cbds 123-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 21704641-8 2011 The correlations between CYP isoform-specific activities and CBD oxidative activities in 16 individual HLMs indicated that 6beta-OH- and 4""-OH-CBDs were mainly formed by CYP3A4, which was supported by inhibition studies using ketoconazole and an anti-CYP3A4 antibody. Ketoconazole 227-239 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 21466844-0 2011 Assessment of a novel beta2-adrenoceptor agonist, trantinterol, for interference with human liver cytochrome P450 enzymes activities. trantinterol 50-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 21757024-1 2011 Arachidonic acid (AA) is metabolized by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes into eicosanoids, which are involved in diverse diseases, including type 1 and type 2 diabetes. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 21757024-1 2011 Arachidonic acid (AA) is metabolized by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes into eicosanoids, which are involved in diverse diseases, including type 1 and type 2 diabetes. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 21757024-1 2011 Arachidonic acid (AA) is metabolized by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) enzymes into eicosanoids, which are involved in diverse diseases, including type 1 and type 2 diabetes. Eicosanoids 121-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 21757024-6 2011 A recent publication has demonstrated that stabilizing the levels of epoxyeicosatrienoic acids (EETs), CYP eicosanoids, by inhibiting or deleting soluble epoxide hydrolase (sEH) improves beta-cell function and reduces beta-cell apoptosis in diabetes. epoxyeicosatrienoic acids 69-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 21757024-6 2011 A recent publication has demonstrated that stabilizing the levels of epoxyeicosatrienoic acids (EETs), CYP eicosanoids, by inhibiting or deleting soluble epoxide hydrolase (sEH) improves beta-cell function and reduces beta-cell apoptosis in diabetes. Eicosanoids 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 21466844-1 2011 The effect of a novel beta(2)-adrenoceptor agonist, trantinterol on the activities of cytochrome P450 (CYP450) was investigated with human liver microsomes and human cryohepatocytes in order to assess the potential for drug-drug interactions. trantinterol 52-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 21466844-1 2011 The effect of a novel beta(2)-adrenoceptor agonist, trantinterol on the activities of cytochrome P450 (CYP450) was investigated with human liver microsomes and human cryohepatocytes in order to assess the potential for drug-drug interactions. trantinterol 52-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-109 21466844-2 2011 The ability of trantinterol to inhibit CYP450 activities was evaluated in vitro in human liver microsomes. trantinterol 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-45 21508826-2 2011 It is metabolized in the liver to norketamine via cytochrome P450 (CYP) enzymes. norketamine 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 21477627-1 2011 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to biologically active eicosanoids. Arachidonic Acid 46-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21477627-1 2011 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to biologically active eicosanoids. Arachidonic Acid 46-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 21477627-1 2011 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to biologically active eicosanoids. Eicosanoids 86-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21477627-1 2011 Cytochrome P450 (CYP) epoxygenases metabolize arachidonic acid to biologically active eicosanoids. Eicosanoids 86-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 21476972-3 2011 However, some CYP enzymes are highly expressed in the heart and catalyze arachidonic acid oxidation to a variety of eicosanoids, which attenuates ischemia-reperfusion injury of the heart. Arachidonic Acid 73-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 21476972-3 2011 However, some CYP enzymes are highly expressed in the heart and catalyze arachidonic acid oxidation to a variety of eicosanoids, which attenuates ischemia-reperfusion injury of the heart. Eicosanoids 116-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 21476972-5 2011 CYP enzymes also represent a significant source of reactive oxygen species (ROS) that may target cellular homeostatic mechanisms and mitochondria. Reactive Oxygen Species 51-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21476972-5 2011 CYP enzymes also represent a significant source of reactive oxygen species (ROS) that may target cellular homeostatic mechanisms and mitochondria. Reactive Oxygen Species 76-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21476972-6 2011 CYP isoforms expressed in the heart are critical for generation of epoxyeicosatrienoic acids (EETs) and ROS. epoxyeicosatrienoic acids 67-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21476972-6 2011 CYP isoforms expressed in the heart are critical for generation of epoxyeicosatrienoic acids (EETs) and ROS. eets 94-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21476972-6 2011 CYP isoforms expressed in the heart are critical for generation of epoxyeicosatrienoic acids (EETs) and ROS. Reactive Oxygen Species 104-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21148022-0 2011 Cytochrome P450 and ABCB1 genetics: association with quetiapine and norquetiapine plasma and cerebrospinal fluid concentrations and with clinical response in patients suffering from schizophrenia. Quetiapine Fumarate 53-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21148022-0 2011 Cytochrome P450 and ABCB1 genetics: association with quetiapine and norquetiapine plasma and cerebrospinal fluid concentrations and with clinical response in patients suffering from schizophrenia. norquetiapine 68-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21148022-4 2011 This study aimed to identify the possible associations of CYP and ABCB1 genetic polymorphisms with quetiapine and norquetiapine plasma and cerebrospinal fluid (CSF) concentrations and with response to treatment. Quetiapine Fumarate 99-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 21148022-4 2011 This study aimed to identify the possible associations of CYP and ABCB1 genetic polymorphisms with quetiapine and norquetiapine plasma and cerebrospinal fluid (CSF) concentrations and with response to treatment. norquetiapine 114-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 21199317-4 2011 We conducted a retrospective analysis of a large commercial claims database and a Medicare database to assess the prevalence of DDEs among patients with osteoarthritis taking CYP450-metabolized opioids. Dichlorodiphenyl Dichloroethylene 128-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-181 21508826-4 2011 The aim of this study was to investigate the effect of CYP enzyme induction by rifampicin on the pharmacokinetics of S-ketamine and its major metabolite, S-norketamine, in healthy volunteers. Ketamine 117-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 21508826-4 2011 The aim of this study was to investigate the effect of CYP enzyme induction by rifampicin on the pharmacokinetics of S-ketamine and its major metabolite, S-norketamine, in healthy volunteers. norketamine 154-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 21508826-2 2011 It is metabolized in the liver to norketamine via cytochrome P450 (CYP) enzymes. norketamine 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 21508826-3 2011 There are few human data on the involvement of CYP enzymes on the elimination of norketamine and its possible contribution to analgesic effect. norketamine 81-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 21508826-4 2011 The aim of this study was to investigate the effect of CYP enzyme induction by rifampicin on the pharmacokinetics of S-ketamine and its major metabolite, S-norketamine, in healthy volunteers. Rifampin 79-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 21463219-7 2011 EXPERT OPINION: This article aims to provide an overview of the multiple gains in incorporating cyclodextrins in poly(anhydride) nanoparticles, including improvement of their bioadhesive capability, the loading of lipophilic drugs and the effect on efflux membrane proteins and cytochrome P450. Cyclodextrins 96-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 278-293 21799712-7 2011 The detection of four cytochrome P-450 enzyme activities was demonstrated following induction by 3-methylcholantlene, with a sensitivity significantly higher than that in conventional monolayer culture. 3-methylcholantlene 97-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 21294626-3 2011 In this study, we characterized the human liver cytochrome P450 (CYP) enzymes responsible for the biotransformation of three major metabolites--M1, M2, and M4--of magnolin. magnolin 163-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 21539368-0 2011 The directive of the protein: how does cytochrome P450 select the mechanism of dopamine formation? Dopamine 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 21539368-1 2011 Dopamine can be generated from tyramine via arene hydroxylation catalyzed by a cytochrome P450 enzyme (CYP2D6). Dopamine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 21539368-1 2011 Dopamine can be generated from tyramine via arene hydroxylation catalyzed by a cytochrome P450 enzyme (CYP2D6). Tyramine 31-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 21539368-1 2011 Dopamine can be generated from tyramine via arene hydroxylation catalyzed by a cytochrome P450 enzyme (CYP2D6). arene 44-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 21543025-4 2011 Cyclophosphamide is metabolized and activated by the cytochrome P450 (CYP) system and in particular CYP enzymes 2B6 and 2C19. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 21543025-4 2011 Cyclophosphamide is metabolized and activated by the cytochrome P450 (CYP) system and in particular CYP enzymes 2B6 and 2C19. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 21543025-4 2011 Cyclophosphamide is metabolized and activated by the cytochrome P450 (CYP) system and in particular CYP enzymes 2B6 and 2C19. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 21504003-6 2011 The suggested reaction pathways included N-methylation leading to iminium formation in primary and/or secondary amines preceded by cytochrome P450 (CYP)-mediated reactions. Nitrogen 41-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 21504003-6 2011 The suggested reaction pathways included N-methylation leading to iminium formation in primary and/or secondary amines preceded by cytochrome P450 (CYP)-mediated reactions. Nitrogen 41-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 21504003-6 2011 The suggested reaction pathways included N-methylation leading to iminium formation in primary and/or secondary amines preceded by cytochrome P450 (CYP)-mediated reactions. iminium 66-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 21504003-6 2011 The suggested reaction pathways included N-methylation leading to iminium formation in primary and/or secondary amines preceded by cytochrome P450 (CYP)-mediated reactions. iminium 66-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 21504003-6 2011 The suggested reaction pathways included N-methylation leading to iminium formation in primary and/or secondary amines preceded by cytochrome P450 (CYP)-mediated reactions. Amines 112-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 21504003-6 2011 The suggested reaction pathways included N-methylation leading to iminium formation in primary and/or secondary amines preceded by cytochrome P450 (CYP)-mediated reactions. Amines 112-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 21112752-0 2011 In vitro effects of natural prenyloxycinnamic acids on human cytochrome P450 isozyme activity and expression. prenyloxycinnamic acids 28-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 21309752-0 2011 Eicosanoid formation by a cytochrome P450 isoform expressed in the pharynx of Caenorhabditis elegans. Eicosanoids 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 21309752-1 2011 Caenorhabditis elegans harbours several CYP (cytochrome P450) genes that are homologous with mammalian CYP isoforms important to the production of physiologically active AA (arachidonic acid) metabolites. Arachidonic Acid 174-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 21309752-1 2011 Caenorhabditis elegans harbours several CYP (cytochrome P450) genes that are homologous with mammalian CYP isoforms important to the production of physiologically active AA (arachidonic acid) metabolites. Arachidonic Acid 174-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 21309752-1 2011 Caenorhabditis elegans harbours several CYP (cytochrome P450) genes that are homologous with mammalian CYP isoforms important to the production of physiologically active AA (arachidonic acid) metabolites. Arachidonic Acid 174-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 21309752-2 2011 We tested the hypothesis that mammals and C. elegans may share similar basic mechanisms of CYP-dependent eicosanoid formation and action. Eicosanoids 105-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 21309752-4 2011 Co-expression of CYP33E2 with the human NADPH-CYP reductase in insect cells resulted in the reconstitution of an active microsomal mono-oxygenase system that metabolized EPA (eicosapentaenoic acid) and, with lower activity, also AA to specific sets of regioisomeric epoxy- and hydroxy-derivatives. Eicosapentaenoic Acid 170-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 21309752-4 2011 Co-expression of CYP33E2 with the human NADPH-CYP reductase in insect cells resulted in the reconstitution of an active microsomal mono-oxygenase system that metabolized EPA (eicosapentaenoic acid) and, with lower activity, also AA to specific sets of regioisomeric epoxy- and hydroxy-derivatives. Eicosapentaenoic Acid 175-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 21309752-8 2011 These results demonstrate that EPA and AA are efficient CYP33E2 substrates and suggest that CYP-eicosanoids, influencing in mammals the contractility of cardiomyocytes and vascular smooth muscle cells, may function in C. elegans as regulators of the pharyngeal pumping activity. Eicosapentaenoic Acid 31-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 21309752-8 2011 These results demonstrate that EPA and AA are efficient CYP33E2 substrates and suggest that CYP-eicosanoids, influencing in mammals the contractility of cardiomyocytes and vascular smooth muscle cells, may function in C. elegans as regulators of the pharyngeal pumping activity. Eicosanoids 96-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 21193036-1 2011 Cytochrome P450 enzymes play an important role in steroid hormone biosynthesis of the human adrenal gland, e.g., the production of cortisol and aldosterone. Steroids 50-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21193036-1 2011 Cytochrome P450 enzymes play an important role in steroid hormone biosynthesis of the human adrenal gland, e.g., the production of cortisol and aldosterone. Hydrocortisone 131-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21193036-1 2011 Cytochrome P450 enzymes play an important role in steroid hormone biosynthesis of the human adrenal gland, e.g., the production of cortisol and aldosterone. Aldosterone 144-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20807350-3 2011 Using a retrospective analysis of a large commercial claims database and a Medicare database, we evaluated DDEs that have the potential to cause DDIs among chronic low back pain (cLBP) patients on long-term opioid analgesia, which metabolizes through the CYP450 enzyme system, concomitant with other CYP450-metabolized drug(s). Dichlorodiphenyl Dichloroethylene 107-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 255-261 20807350-3 2011 Using a retrospective analysis of a large commercial claims database and a Medicare database, we evaluated DDEs that have the potential to cause DDIs among chronic low back pain (cLBP) patients on long-term opioid analgesia, which metabolizes through the CYP450 enzyme system, concomitant with other CYP450-metabolized drug(s). Dichlorodiphenyl Dichloroethylene 107-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 300-306 21210720-11 2011 Drug-induced cytochrome P450 gene expression was increased with rifampicin induction. Rifampin 64-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 21294626-3 2011 In this study, we characterized the human liver cytochrome P450 (CYP) enzymes responsible for the biotransformation of three major metabolites--M1, M2, and M4--of magnolin. magnolin 163-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 21294626-4 2011 CYP2C8, CYP2C9, CYP2C19, and CYP3A4 were identified as the major enzymes responsible for the formation of the two O-desmethyl magnolins (M1 and M2), on the basis of a combination of correlation analysis and experiments, including immunoinhibition of magnolin in human liver microsomes and metabolism of magnolin by human cDNA-expressed CYP enzymes. magnolin 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21294626-4 2011 CYP2C8, CYP2C9, CYP2C19, and CYP3A4 were identified as the major enzymes responsible for the formation of the two O-desmethyl magnolins (M1 and M2), on the basis of a combination of correlation analysis and experiments, including immunoinhibition of magnolin in human liver microsomes and metabolism of magnolin by human cDNA-expressed CYP enzymes. magnolin 250-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21444768-0 2011 Understanding the determinants of selectivity in drug metabolism through modeling of dextromethorphan oxidation by cytochrome P450. Dextromethorphan 85-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-130 21363914-4 2011 Here, we use a new mathematical model to compare the effects of conventional cyclophosphamide therapy with those induced when macrophages are used to deliver hypoxia-inducible cytochrome P450 to locally activate cyclophosphamide. Cyclophosphamide 77-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-191 21363914-4 2011 Here, we use a new mathematical model to compare the effects of conventional cyclophosphamide therapy with those induced when macrophages are used to deliver hypoxia-inducible cytochrome P450 to locally activate cyclophosphamide. Cyclophosphamide 212-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-191 20851588-12 2011 The results provide initial structure-activity information about the interaction of isoquinoline alkaloids with major human xenobiotic-metabolizing CYP enzymes, and illustrate potential novel structures as CYP form-selective inhibitors. isoquinoline alkaloids 84-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 20851588-12 2011 The results provide initial structure-activity information about the interaction of isoquinoline alkaloids with major human xenobiotic-metabolizing CYP enzymes, and illustrate potential novel structures as CYP form-selective inhibitors. isoquinoline alkaloids 84-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-209 21348461-3 2011 Here we show that the selective cytotoxic action of austocystin D arises from its selective activation by cytochrome P450 (CYP) enzymes in specific cancer cell lines, leading to induction of DNA damage in cells and in vitro. austocystin D 52-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 21348461-3 2011 Here we show that the selective cytotoxic action of austocystin D arises from its selective activation by cytochrome P450 (CYP) enzymes in specific cancer cell lines, leading to induction of DNA damage in cells and in vitro. austocystin D 52-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 21348461-5 2011 Furthermore, the pattern of cytotoxicity of austocystin D was distinct from doxorubicin and etoposide and unlike aflatoxin B(1), a compound that resembles austocystin D and is also activated by CYP enzymes to induce DNA damage. austocystin D 44-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-197 20851588-0 2011 Inhibition of human drug metabolizing cytochrome P450 enzymes by plant isoquinoline alkaloids. isoquinoline alkaloids 71-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 20851588-3 2011 The purpose of this study was to determine in vitro CYP inhibition potencies of a set of isoquinoline alkaloids to gain insight into interactions of novel chemical structures with CYP enzymes. isoquinoline alkaloids 89-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 20851588-3 2011 The purpose of this study was to determine in vitro CYP inhibition potencies of a set of isoquinoline alkaloids to gain insight into interactions of novel chemical structures with CYP enzymes. isoquinoline alkaloids 89-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 21392639-2 2011 Clopidogrel, a prodrug, requires hepatic cytochrome P450 (CYP) metabolic activation to produce the active metabolite that inhibits the platelet P2Y12 adenosine diphosphate (ADP) receptor, decreasing platelet activation and aggregation processes. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 21276781-11 2011 These findings suggest that co-administration of OS products, especially those with high eupatorin content, with conventional drugs may have the potential to cause drug-herb interactions involving inhibition of major CYP enzymes. eupatorin 89-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 217-220 20484614-8 2011 The increase in tPDE4i of roflumilast and roflumilast N-oxide following coadministration with cimetidine was mainly due to the inhibitory effect of cimetidine on cytochrome P450 (CYP) isoenzymes CYP1A2, CYP3A, and CYP2C19. n-oxide 54-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 20484614-8 2011 The increase in tPDE4i of roflumilast and roflumilast N-oxide following coadministration with cimetidine was mainly due to the inhibitory effect of cimetidine on cytochrome P450 (CYP) isoenzymes CYP1A2, CYP3A, and CYP2C19. n-oxide 54-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-182 20484614-8 2011 The increase in tPDE4i of roflumilast and roflumilast N-oxide following coadministration with cimetidine was mainly due to the inhibitory effect of cimetidine on cytochrome P450 (CYP) isoenzymes CYP1A2, CYP3A, and CYP2C19. Cimetidine 94-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 20484614-8 2011 The increase in tPDE4i of roflumilast and roflumilast N-oxide following coadministration with cimetidine was mainly due to the inhibitory effect of cimetidine on cytochrome P450 (CYP) isoenzymes CYP1A2, CYP3A, and CYP2C19. Cimetidine 94-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-182 20484614-8 2011 The increase in tPDE4i of roflumilast and roflumilast N-oxide following coadministration with cimetidine was mainly due to the inhibitory effect of cimetidine on cytochrome P450 (CYP) isoenzymes CYP1A2, CYP3A, and CYP2C19. Cimetidine 148-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 20484614-8 2011 The increase in tPDE4i of roflumilast and roflumilast N-oxide following coadministration with cimetidine was mainly due to the inhibitory effect of cimetidine on cytochrome P450 (CYP) isoenzymes CYP1A2, CYP3A, and CYP2C19. Cimetidine 148-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-182 21392639-3 2011 Atorvastatin, omeprazole, and several other drugs have been shown in pharmacodynamic studies to competitively inhibit CYP activation of clopidogrel, reducing clopidogrel responsiveness. Omeprazole 14-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 21392639-2 2011 Clopidogrel, a prodrug, requires hepatic cytochrome P450 (CYP) metabolic activation to produce the active metabolite that inhibits the platelet P2Y12 adenosine diphosphate (ADP) receptor, decreasing platelet activation and aggregation processes. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 21392639-3 2011 Atorvastatin, omeprazole, and several other drugs have been shown in pharmacodynamic studies to competitively inhibit CYP activation of clopidogrel, reducing clopidogrel responsiveness. Clopidogrel 136-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 21392639-3 2011 Atorvastatin, omeprazole, and several other drugs have been shown in pharmacodynamic studies to competitively inhibit CYP activation of clopidogrel, reducing clopidogrel responsiveness. Clopidogrel 158-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 21392639-3 2011 Atorvastatin, omeprazole, and several other drugs have been shown in pharmacodynamic studies to competitively inhibit CYP activation of clopidogrel, reducing clopidogrel responsiveness. Atorvastatin 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 21392639-4 2011 Conversely, other agents increase clopidogrel responsiveness by inducing CYP activity. Clopidogrel 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 49-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-174 21356152-0 2011 Cytochrome P450 eicosanoids and cerebral vascular function. Eicosanoids 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. Eicosanoids 4-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-174 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. Eicosanoids 4-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-179 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 16-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-174 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 16-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-179 21393488-10 2011 Caution should be exercised when coadministering any statin with drugs that metabolize through cytochrome P-450 IIIA-4 in particular fibrates, cyclosporine, and azole antifungals. Cyclosporine 143-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 20951188-1 2011 Cytochrome P450 (CYP) enzyme inhibitory properties of six chromenylated amide compounds (CAs) from Amyris plumieri are described. Amides 72-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20951188-1 2011 Cytochrome P450 (CYP) enzyme inhibitory properties of six chromenylated amide compounds (CAs) from Amyris plumieri are described. Amides 72-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 20951188-1 2011 Cytochrome P450 (CYP) enzyme inhibitory properties of six chromenylated amide compounds (CAs) from Amyris plumieri are described. cas 89-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20951188-1 2011 Cytochrome P450 (CYP) enzyme inhibitory properties of six chromenylated amide compounds (CAs) from Amyris plumieri are described. cas 89-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 49-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-179 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. epoxyeicosatrienoic acids 62-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-174 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. epoxyeicosatrienoic acids 62-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-179 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. eets 89-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-174 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. eets 89-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-179 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. Arachidonic Acid 139-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-174 21356152-1 2011 The eicosanoids 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs), which are generated from the metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes, possess a wide array of biological actions, including the regulation of blood flow to organs. Arachidonic Acid 139-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-179 21356152-3 2011 Investigations are beginning to unravel the molecular and cellular mechanisms by which these CYP eicosanoids regulate cerebral vascular function and the changes that occur in pathological states. Eicosanoids 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 21356152-7 2011 Thus, the CYP eicosanoids are key regulators of cerebral vascular function and novel therapeutic targets for cardiovascular diseases and neurological disorders. Eicosanoids 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-13 20723094-2 2011 BACKGROUND: Tramadol (M) is metabolized by O-demethylation (cytochrome P450 [CYP] 2D6) to the pharmacodynamic active metabolite O-demethyl tramadol (M1). Tramadol 12-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-85 20845077-2 2011 Cytochrome P450 (CYP) polymorphisms have been proposed as possible mechanisms for nonresponsiveness to clopidogrel. Clopidogrel 103-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20845077-2 2011 Cytochrome P450 (CYP) polymorphisms have been proposed as possible mechanisms for nonresponsiveness to clopidogrel. Clopidogrel 103-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 20723094-2 2011 BACKGROUND: Tramadol (M) is metabolized by O-demethylation (cytochrome P450 [CYP] 2D6) to the pharmacodynamic active metabolite O-demethyl tramadol (M1). O-demethyltramadol 128-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-85 21401511-5 2011 These two dopaminergic pathways stimulate the secretion of pituitary hormones, which directly (GH) or indirectly (ACTH, TSH) activate hepatic nuclear/ cytosolic receptors controlling CYP genes. Thyrotropin 120-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-186 21147774-1 2011 Cytochrome P450 (P450) 7A1 is well known as the cholesterol 7alpha-hydroxylase, the first enzyme involved in bile acid synthesis from cholesterol. Bile Acids and Salts 109-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 21147774-1 2011 Cytochrome P450 (P450) 7A1 is well known as the cholesterol 7alpha-hydroxylase, the first enzyme involved in bile acid synthesis from cholesterol. Cholesterol 48-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 26596155-1 2011 The mixed density functional theory (DFT) and valence bond study described herein focuses on the activation of 17 benzene derivatives by the active species of Cytochrome P450, so-called Compound I (Cpd I), as well as by the methoxy radical, as a potentially simple model of Cpd I (Jones, J. P.; Mysinger, M.; Korzekwa, K. R. Drug Metab. Benzene 114-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-174 21401511-6 2011 Recent preliminary studies with selective noradrenaline or serotonin neurotoxins suggest also involvement of the brain noradrenergic and serotonergic systems in the regulation of liver CYP. Norepinephrine 42-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-188 21401511-7 2011 Moreover, the influence of the peripheral nervous system involving several neurotransmitters (acetylcholine, noradrenaline, adrenaline, dopamine, serotonin) on liver function may also be important for the physiological regulation of hepatic CYP activity. Acetylcholine 94-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-244 20860521-0 2011 Cytochrome P450 regulation: the interplay between its heme and apoprotein moieties in synthesis, assembly, repair, and disposal. Heme 54-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21401511-7 2011 Moreover, the influence of the peripheral nervous system involving several neurotransmitters (acetylcholine, noradrenaline, adrenaline, dopamine, serotonin) on liver function may also be important for the physiological regulation of hepatic CYP activity. Norepinephrine 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-244 21401511-7 2011 Moreover, the influence of the peripheral nervous system involving several neurotransmitters (acetylcholine, noradrenaline, adrenaline, dopamine, serotonin) on liver function may also be important for the physiological regulation of hepatic CYP activity. Epinephrine 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-244 21401511-7 2011 Moreover, the influence of the peripheral nervous system involving several neurotransmitters (acetylcholine, noradrenaline, adrenaline, dopamine, serotonin) on liver function may also be important for the physiological regulation of hepatic CYP activity. Dopamine 136-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-244 21401511-7 2011 Moreover, the influence of the peripheral nervous system involving several neurotransmitters (acetylcholine, noradrenaline, adrenaline, dopamine, serotonin) on liver function may also be important for the physiological regulation of hepatic CYP activity. Serotonin 146-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-244 21087116-1 2011 This study aims to characterize the metabolism of alpha-thujone in human liver preparations in vitro and to identify the role of cytochrome P450 (CYP) and possibly other enzymes catalyzing alpha-thujone biotransformations. beta-thujone 189-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-144 21068193-0 2011 Inactivation of cytochrome P450 (P450) 3A4 but not P450 3A5 by OSI-930, a thiophene-containing anticancer drug. OSI 930 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-42 21068193-0 2011 Inactivation of cytochrome P450 (P450) 3A4 but not P450 3A5 by OSI-930, a thiophene-containing anticancer drug. Thiophenes 74-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-42 21068193-2 2011 Results showed that OSI-930 inactivated purified, recombinant cytochrome P450 (P450) 3A4 in the reconstituted system in a mechanism-based manner. OSI 930 20-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-88 20641061-0 2011 Identification of cytochrome P450 (CYP) isoforms involved in the metabolism of corynoline, and assessment of its herb-drug interactions. corynoline 79-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 20641061-0 2011 Identification of cytochrome P450 (CYP) isoforms involved in the metabolism of corynoline, and assessment of its herb-drug interactions. corynoline 79-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 20641061-5 2011 Among seven major CYP isoforms tested, corynoline showed strong inhibitory effects on the activities of CYP3A4 and CYP2C9, with an IC(50) of 3.3 +- 0.9 microm and 31.5 +- 0.5 microm, respectively. corynoline 39-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 21087116-1 2011 This study aims to characterize the metabolism of alpha-thujone in human liver preparations in vitro and to identify the role of cytochrome P450 (CYP) and possibly other enzymes catalyzing alpha-thujone biotransformations. beta-thujone 189-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 21087116-6 2011 Screening of alpha-thujone metabolism with CYP recombinant enzymes indicated that CYP2A6 was principally responsible for the major 7- and 4-hydroxylation reactions, although CYP3A4 and CYP2B6 participated to a lesser extent and CYP3A4 and CYP2B6 catalyzed minor 2-hydroxylation. beta-thujone 13-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 21251580-2 2011 BACKGROUND: CCBs inhibit a variety of cytochrome P-450 enzymes, some of which contribute to clopidogrel metabolic activation. Clopidogrel 92-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 21168332-0 2011 (13)C-labeled indolequinone-DTPA-Gd conjugate for NMR probing cytochrome:P450 reductase-mediated one-electron reduction. Indolequinones 14-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 21168546-2 2011 The enzyme cytochrome P450 was covalently attached to screen-printed carbon electrodes. Carbon 69-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-26 20825389-1 2011 In vitro metabolism of methadone was investigated in cytochrome P450 (CYP) supersomes and phenotyped human liver microsomes (HLMs) to reconcile past findings on CYP involvement in stereo-selective metabolism of methadone. Methadone 23-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 21039635-3 2011 This study is designed to assess the effect of alcohol on the ABCC1 and CYP enzymes involved in the metabolism of NNRTIs and PIs (CYP2B6, CYP2D6, and CYP3A4) and oxidative stress (CYP1A1, CYP2A6, and CYP2E1) in U937 macrophages. Alcohols 47-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 21226615-2 2011 The conversion of tamoxifen to active metabolites involves several cytochrome P450 (CYP) enzymes. Tamoxifen 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 21226615-2 2011 The conversion of tamoxifen to active metabolites involves several cytochrome P450 (CYP) enzymes. Tamoxifen 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 22126492-2 2011 Many CYP enzymes function in the liver, but presence of CYP2E1 in the brain is demonstrating its role in both nicotine and ethanol metabolism. Ethanol 123-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-8 21168332-0 2011 (13)C-labeled indolequinone-DTPA-Gd conjugate for NMR probing cytochrome:P450 reductase-mediated one-electron reduction. Pentetic Acid 28-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 21168332-0 2011 (13)C-labeled indolequinone-DTPA-Gd conjugate for NMR probing cytochrome:P450 reductase-mediated one-electron reduction. Gadolinium 33-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 21168332-1 2011 We designed and synthesized a new class of (13)C-labeled NMR probe, (13)C-IQ-Gd, to monitor one-electron reductions by cytochrome:P450 (CYP450) reductase under hypoxic conditions. Carbon 47-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-134 21168332-1 2011 We designed and synthesized a new class of (13)C-labeled NMR probe, (13)C-IQ-Gd, to monitor one-electron reductions by cytochrome:P450 (CYP450) reductase under hypoxic conditions. Carbon 47-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-142 21168332-3 2011 The (13)C NMR signal of (13)C-IQ-Gd was suppressed because of the intramolecular paramagnetic effect of Gd(3+), whereas enzymatic reduction mediated by CYP450 reductase under hypoxic conditions yielded an intensed (13)C NMR signal due to enzymatic activation of the IQ unit followed by release of the DTPA-Gd unit from (13)C-IQ-Gd. Pentetic Acid 301-305 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-158 21039635-11 2011 CONCLUSIONS: Our study showed that alcohol causes increases in the genetic and functional expressions of ABCC1 and CYP enzymes in U937 macrophages. Alcohols 35-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 20825389-1 2011 In vitro metabolism of methadone was investigated in cytochrome P450 (CYP) supersomes and phenotyped human liver microsomes (HLMs) to reconcile past findings on CYP involvement in stereo-selective metabolism of methadone. Methadone 23-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 20825389-1 2011 In vitro metabolism of methadone was investigated in cytochrome P450 (CYP) supersomes and phenotyped human liver microsomes (HLMs) to reconcile past findings on CYP involvement in stereo-selective metabolism of methadone. Methadone 23-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 20825389-1 2011 In vitro metabolism of methadone was investigated in cytochrome P450 (CYP) supersomes and phenotyped human liver microsomes (HLMs) to reconcile past findings on CYP involvement in stereo-selective metabolism of methadone. Methadone 211-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 20825389-3 2011 CYP supersome activity for methadone use and EDDP formation ranked CYP2B6 > 3A4 > 2C19 > 2D6 > 2C18, 3A7 > 2C8, 2C9, 3A5. Methadone 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 21532165-0 2011 In vitro characterization of the cytochrome P450 isoforms involved in the metabolism of 6-methoxy-2-napthylacetic acid, an active metabolite of the prodrug nabumetone. 6-methoxy-2-napthylacetic acid 88-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 20576524-3 2011 The CYP-mediated ellipticine metabolites 9-hydroxy- and 7-hydroxyellipticine and the product of ellipticine oxidation by peroxidases, the ellipticine dimer, are the detoxication metabolites of this compound. ellipticine 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 20869469-0 2011 Role of cytochrome P450 enzymes in the bioactivation of polyunsaturated fatty acids. Fatty Acids, Unsaturated 56-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 20869469-1 2011 Cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA), such as epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid, serve as second messengers of various hormones and growth factors and play pivotal roles in the regulation of vascular, renal and cardiac function. Arachidonic Acid 47-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20869469-1 2011 Cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA), such as epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid, serve as second messengers of various hormones and growth factors and play pivotal roles in the regulation of vascular, renal and cardiac function. Arachidonic Acid 47-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 20869469-1 2011 Cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA), such as epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid, serve as second messengers of various hormones and growth factors and play pivotal roles in the regulation of vascular, renal and cardiac function. epoxyeicosatrienoic acids 78-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20869469-1 2011 Cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA), such as epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid, serve as second messengers of various hormones and growth factors and play pivotal roles in the regulation of vascular, renal and cardiac function. epoxyeicosatrienoic acids 78-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 20869469-1 2011 Cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA), such as epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid, serve as second messengers of various hormones and growth factors and play pivotal roles in the regulation of vascular, renal and cardiac function. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 108-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20869469-1 2011 Cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA), such as epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid, serve as second messengers of various hormones and growth factors and play pivotal roles in the regulation of vascular, renal and cardiac function. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 108-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 20869469-2 2011 As discussed in the present review, virtually all of the major AA metabolizing CYP isoforms accept a variety of other polyunsaturated fatty acids (PUFA), including linoleic, eicosapentaenoic (EPA) and docosahexaenoic acids (DHA), as efficient alternative substrates. Fatty Acids, Unsaturated 118-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 20869469-2 2011 As discussed in the present review, virtually all of the major AA metabolizing CYP isoforms accept a variety of other polyunsaturated fatty acids (PUFA), including linoleic, eicosapentaenoic (EPA) and docosahexaenoic acids (DHA), as efficient alternative substrates. Fatty Acids, Unsaturated 147-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 20869469-2 2011 As discussed in the present review, virtually all of the major AA metabolizing CYP isoforms accept a variety of other polyunsaturated fatty acids (PUFA), including linoleic, eicosapentaenoic (EPA) and docosahexaenoic acids (DHA), as efficient alternative substrates. Linoleic Acid 164-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 20869469-2 2011 As discussed in the present review, virtually all of the major AA metabolizing CYP isoforms accept a variety of other polyunsaturated fatty acids (PUFA), including linoleic, eicosapentaenoic (EPA) and docosahexaenoic acids (DHA), as efficient alternative substrates. eicosapentaenoic 174-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 20869469-2 2011 As discussed in the present review, virtually all of the major AA metabolizing CYP isoforms accept a variety of other polyunsaturated fatty acids (PUFA), including linoleic, eicosapentaenoic (EPA) and docosahexaenoic acids (DHA), as efficient alternative substrates. Eicosapentaenoic Acid 192-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 20869469-2 2011 As discussed in the present review, virtually all of the major AA metabolizing CYP isoforms accept a variety of other polyunsaturated fatty acids (PUFA), including linoleic, eicosapentaenoic (EPA) and docosahexaenoic acids (DHA), as efficient alternative substrates. Docosahexaenoic Acids 201-222 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 20869469-2 2011 As discussed in the present review, virtually all of the major AA metabolizing CYP isoforms accept a variety of other polyunsaturated fatty acids (PUFA), including linoleic, eicosapentaenoic (EPA) and docosahexaenoic acids (DHA), as efficient alternative substrates. Docosahexaenoic Acids 224-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 20869469-7 2011 Taken together, these findings indicate that CYP-dependent signaling pathways are highly susceptible to changes in the relative bioavailability of the different PUFAs and may provide novel insight into the complex mechanisms that link essential dietary fatty acids to the development of cardiovascular disease. essential dietary fatty acids 235-264 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 20576524-0 2011 Cytochrome P450- and peroxidase-mediated oxidation of anticancer alkaloid ellipticine dictates its anti-tumor efficiency. Alkaloids 65-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-31 20576524-0 2011 Cytochrome P450- and peroxidase-mediated oxidation of anticancer alkaloid ellipticine dictates its anti-tumor efficiency. ellipticine 74-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-31 20576524-1 2011 An antineoplastic alkaloid ellipticine is a prodrug, whose pharmacological efficiency is dependent on its cytochrome P450 (CYP)- and/or peroxidase-mediated activation in target tissues. Alkaloids 18-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 20576524-1 2011 An antineoplastic alkaloid ellipticine is a prodrug, whose pharmacological efficiency is dependent on its cytochrome P450 (CYP)- and/or peroxidase-mediated activation in target tissues. Alkaloids 18-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 20576524-1 2011 An antineoplastic alkaloid ellipticine is a prodrug, whose pharmacological efficiency is dependent on its cytochrome P450 (CYP)- and/or peroxidase-mediated activation in target tissues. ellipticine 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 20576524-1 2011 An antineoplastic alkaloid ellipticine is a prodrug, whose pharmacological efficiency is dependent on its cytochrome P450 (CYP)- and/or peroxidase-mediated activation in target tissues. ellipticine 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 20576524-3 2011 The CYP-mediated ellipticine metabolites 9-hydroxy- and 7-hydroxyellipticine and the product of ellipticine oxidation by peroxidases, the ellipticine dimer, are the detoxication metabolites of this compound. ellipticine 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 20576524-3 2011 The CYP-mediated ellipticine metabolites 9-hydroxy- and 7-hydroxyellipticine and the product of ellipticine oxidation by peroxidases, the ellipticine dimer, are the detoxication metabolites of this compound. 9-hydroxy- and 7-hydroxyellipticine 41-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 20576524-3 2011 The CYP-mediated ellipticine metabolites 9-hydroxy- and 7-hydroxyellipticine and the product of ellipticine oxidation by peroxidases, the ellipticine dimer, are the detoxication metabolites of this compound. ellipticine 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 20576524-8 2011 It also suggests ellipticine reactive metabolites 13-hydroxyellipticine and 12-hydroxyellipticine to be good candidates for targeting to tumors absent from the CYP and peroxidase activation enzymes. ellipticine 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-163 20576524-8 2011 It also suggests ellipticine reactive metabolites 13-hydroxyellipticine and 12-hydroxyellipticine to be good candidates for targeting to tumors absent from the CYP and peroxidase activation enzymes. 13-hydroxyellipticine 50-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-163 20576524-8 2011 It also suggests ellipticine reactive metabolites 13-hydroxyellipticine and 12-hydroxyellipticine to be good candidates for targeting to tumors absent from the CYP and peroxidase activation enzymes. 12-hydroxyellipticine 76-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-163 21532165-0 2011 In vitro characterization of the cytochrome P450 isoforms involved in the metabolism of 6-methoxy-2-napthylacetic acid, an active metabolite of the prodrug nabumetone. Nabumetone 156-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 21198441-2 2011 The specific CYP substrates used in this cocktail assay included phenacetin (CYP1A2), bupropion (CYP2B6), amodiaquine (CYP2C8), tolbutamide (CYP2C9), S-mephenytoin (CYP2C19), dextromethorphan (CYP2D6), and midazolam (CYP3A4/5). Phenacetin 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 21198441-2 2011 The specific CYP substrates used in this cocktail assay included phenacetin (CYP1A2), bupropion (CYP2B6), amodiaquine (CYP2C8), tolbutamide (CYP2C9), S-mephenytoin (CYP2C19), dextromethorphan (CYP2D6), and midazolam (CYP3A4/5). Mephenytoin 150-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 21084761-1 2011 Cytochrome P450 oxidoreductase (POR) transfers electrons from NADPH to all microsomal cytochrome P450 (CYP) enzymes and is necessary for microsomal CYP activities. NADP 62-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 21084761-1 2011 Cytochrome P450 oxidoreductase (POR) transfers electrons from NADPH to all microsomal cytochrome P450 (CYP) enzymes and is necessary for microsomal CYP activities. NADP 62-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 21084761-1 2011 Cytochrome P450 oxidoreductase (POR) transfers electrons from NADPH to all microsomal cytochrome P450 (CYP) enzymes and is necessary for microsomal CYP activities. NADP 62-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 21198441-2 2011 The specific CYP substrates used in this cocktail assay included phenacetin (CYP1A2), bupropion (CYP2B6), amodiaquine (CYP2C8), tolbutamide (CYP2C9), S-mephenytoin (CYP2C19), dextromethorphan (CYP2D6), and midazolam (CYP3A4/5). Dextromethorphan 175-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 21198441-2 2011 The specific CYP substrates used in this cocktail assay included phenacetin (CYP1A2), bupropion (CYP2B6), amodiaquine (CYP2C8), tolbutamide (CYP2C9), S-mephenytoin (CYP2C19), dextromethorphan (CYP2D6), and midazolam (CYP3A4/5). Midazolam 206-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 21771377-3 2011 Concomitant use of omeprazole and clopidogrel was found to decrease the exposure (AUC) to clopidogrel"s active metabolite by 50% and to sharply increase platelet reactivity, as a result of inhibition by omeprazole of CYP2C19, a cytochrome P450 (CYP) enzyme. Omeprazole 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 21205193-0 2011 Cytochrome P450 metabolizing fatty acids in living organisms. Fatty Acids 29-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21047828-1 2011 BACKGROUND: rifampicin lowers nevirapine plasma concentrations by inducing cytochrome P450. Rifampin 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 21047828-1 2011 BACKGROUND: rifampicin lowers nevirapine plasma concentrations by inducing cytochrome P450. Nevirapine 30-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 20980920-1 2011 Clopidogrel is metabolically activated by cytochrome P450 (CYP) isoenzymes. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 20980920-1 2011 Clopidogrel is metabolically activated by cytochrome P450 (CYP) isoenzymes. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 21142269-0 2011 Effect of inhibition of cytochrome P450 enzymes 2D6 and 3A4 on the pharmacokinetics of intravenous oxycodone: a randomized, three-phase, crossover, placebo-controlled study. Oxycodone 99-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 21142269-1 2011 BACKGROUND AND OBJECTIVE: Oxycodone is a mu-opioid receptor agonist that is mainly metabolized by hepatic cytochrome P450 (CYP) enzymes. Oxycodone 26-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 21142269-1 2011 BACKGROUND AND OBJECTIVE: Oxycodone is a mu-opioid receptor agonist that is mainly metabolized by hepatic cytochrome P450 (CYP) enzymes. Oxycodone 26-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 21142269-2 2011 Because CYP enzymes can be inhibited by other drugs, the pharmacokinetics of oxycodone are prone to drug interactions. Oxycodone 77-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-11 21574905-9 2011 CONCLUSION: Since concurrent exposure to DDEs with the potential to cause PK DDIs may be relatively common, policy decisions-makers should consider the use of long-acting opioids that are not metabolized through the CYP450 pathway. Dichlorodiphenyl Dichloroethylene 41-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-222 21771377-3 2011 Concomitant use of omeprazole and clopidogrel was found to decrease the exposure (AUC) to clopidogrel"s active metabolite by 50% and to sharply increase platelet reactivity, as a result of inhibition by omeprazole of CYP2C19, a cytochrome P450 (CYP) enzyme. Omeprazole 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 217-220 21771377-3 2011 Concomitant use of omeprazole and clopidogrel was found to decrease the exposure (AUC) to clopidogrel"s active metabolite by 50% and to sharply increase platelet reactivity, as a result of inhibition by omeprazole of CYP2C19, a cytochrome P450 (CYP) enzyme. Clopidogrel 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-243 21771377-3 2011 Concomitant use of omeprazole and clopidogrel was found to decrease the exposure (AUC) to clopidogrel"s active metabolite by 50% and to sharply increase platelet reactivity, as a result of inhibition by omeprazole of CYP2C19, a cytochrome P450 (CYP) enzyme. Clopidogrel 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 217-220 21558760-1 2011 This review focuses on the development of ruthenium and flavin catalysts for environmentally benign oxidation reactions based on mimicking the functions of cytochrome P-450 and flavoenzymes, and low valent transition-metal catalysts that replace conventional acids and bases. Ruthenium 42-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 21558760-1 2011 This review focuses on the development of ruthenium and flavin catalysts for environmentally benign oxidation reactions based on mimicking the functions of cytochrome P-450 and flavoenzymes, and low valent transition-metal catalysts that replace conventional acids and bases. 4,6-dinitro-o-cresol 56-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 21228620-1 2011 Talarozole is a new highly potent and selective azole derivative which inhibits cytochrome-P450-dependent all-trans-retinoic acid catabolism. R 115866 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 21546883-2 2011 The thienopyridine clopidogrel is a prodrug that requires bioactivation by the cytochrome P450 (CYP) system in order to exert its antiplatelet effect. thienopyridine 4-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 21546883-2 2011 The thienopyridine clopidogrel is a prodrug that requires bioactivation by the cytochrome P450 (CYP) system in order to exert its antiplatelet effect. thienopyridine 4-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 21546883-2 2011 The thienopyridine clopidogrel is a prodrug that requires bioactivation by the cytochrome P450 (CYP) system in order to exert its antiplatelet effect. Clopidogrel 19-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 21546883-2 2011 The thienopyridine clopidogrel is a prodrug that requires bioactivation by the cytochrome P450 (CYP) system in order to exert its antiplatelet effect. Clopidogrel 19-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 21228620-1 2011 Talarozole is a new highly potent and selective azole derivative which inhibits cytochrome-P450-dependent all-trans-retinoic acid catabolism. Azoles 48-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 21228620-1 2011 Talarozole is a new highly potent and selective azole derivative which inhibits cytochrome-P450-dependent all-trans-retinoic acid catabolism. Tretinoin 110-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 21098752-13 2010 Cytochrome P450 enzyme induction by phenobarbital could be responsible for the production of reactive metabolites of lamotrigine that might be causative for the observed hematologic effects. Phenobarbital 36-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21462897-0 2011 In vitro metabolism of glycyrrhetic acid by human cytochrome P450. Glycyrrhetinic Acid 23-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 20609441-0 2010 Enantioselective capillary electrophoresis for identification and characterization of human cytochrome P450 enzymes which metabolize ketamine and norketamine in vitro. Ketamine 133-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 20609441-0 2010 Enantioselective capillary electrophoresis for identification and characterization of human cytochrome P450 enzymes which metabolize ketamine and norketamine in vitro. norketamine 146-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 20609441-3 2010 Enantioselective capillary electrophoresis with multiple isomer sulfated beta-cyclodextrin as chiral selector was used to identify cytochrome P450 enzymes involved in hepatic ketamine and norketamine biotransformation in vitro. betadex 73-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 20609441-3 2010 Enantioselective capillary electrophoresis with multiple isomer sulfated beta-cyclodextrin as chiral selector was used to identify cytochrome P450 enzymes involved in hepatic ketamine and norketamine biotransformation in vitro. Ketamine 175-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 20609441-3 2010 Enantioselective capillary electrophoresis with multiple isomer sulfated beta-cyclodextrin as chiral selector was used to identify cytochrome P450 enzymes involved in hepatic ketamine and norketamine biotransformation in vitro. norketamine 188-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 20609441-4 2010 The N-demethylation of ketamine to norketamine and subsequently the biotransformation of norketamine to other metabolites were studied via analysis of alkaline extracts of in vitro incubations of racemic ketamine and racemic norketamine with nine recombinantly expressed human cytochrome P450 enzymes and human liver microsomes. Ketamine 23-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 277-292 21098752-13 2010 Cytochrome P450 enzyme induction by phenobarbital could be responsible for the production of reactive metabolites of lamotrigine that might be causative for the observed hematologic effects. Lamotrigine 117-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20941528-0 2010 Cytochrome P450-derived eicosanoids: the neglected pathway in cancer. Eicosanoids 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 21191764-3 2010 This study characterized the human liver cytochrome P450 (CYP) and UDPglucuronosyltransferase (UGT) enzymes responsible for the metabolism of jaceosidin. jaceosidin 142-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 21191764-3 2010 This study characterized the human liver cytochrome P450 (CYP) and UDPglucuronosyltransferase (UGT) enzymes responsible for the metabolism of jaceosidin. jaceosidin 142-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 21191764-4 2010 CYP1A2 was identified as the major enzyme responsible for the formation of 6-O-desmethyljaceosidin and hydroxyjaceosidin from jaceosidin on the basis of a combination of correlation analysis and experiments including immuno-inhibition, chemical inhibition in human liver microsomes, and metabolism by human cDNA-expressed CYP enzymes. 6-o-desmethyljaceosidin 75-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 20941528-4 2010 However, arachidonic acid is also substrate for a third enzymatic pathway, the cytochrome P450 (CYP) system. Arachidonic Acid 9-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 21191764-4 2010 CYP1A2 was identified as the major enzyme responsible for the formation of 6-O-desmethyljaceosidin and hydroxyjaceosidin from jaceosidin on the basis of a combination of correlation analysis and experiments including immuno-inhibition, chemical inhibition in human liver microsomes, and metabolism by human cDNA-expressed CYP enzymes. hydroxyjaceosidin 103-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 20941528-4 2010 However, arachidonic acid is also substrate for a third enzymatic pathway, the cytochrome P450 (CYP) system. Arachidonic Acid 9-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 21191764-4 2010 CYP1A2 was identified as the major enzyme responsible for the formation of 6-O-desmethyljaceosidin and hydroxyjaceosidin from jaceosidin on the basis of a combination of correlation analysis and experiments including immuno-inhibition, chemical inhibition in human liver microsomes, and metabolism by human cDNA-expressed CYP enzymes. jaceosidin 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 20844025-2 2010 This is due to disruption of electron transfer from mutant POR to microsomal cytochrome P450 (CYP) enzymes that play a key role in glucocorticoid and sex steroid synthesis. Steroids 154-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 24688155-1 2010 BACKGROUND: Fluoxetine is an inhibitor of the main metabolizing enzymes (cytochrome P450 [CYP] 2C19 and CYP3A4) of omeprazole and thus might influence that drug"s pharmacokinetics. Fluoxetine 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-99 24688155-1 2010 BACKGROUND: Fluoxetine is an inhibitor of the main metabolizing enzymes (cytochrome P450 [CYP] 2C19 and CYP3A4) of omeprazole and thus might influence that drug"s pharmacokinetics. Omeprazole 115-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-99 20851877-0 2010 Metabolism of sesamin by cytochrome P450 in human liver microsomes. sesamin 14-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 20851877-1 2010 Metabolism of sesamin by cytochrome P450 (P450) was examined using yeast expression system and human liver microsomes. sesamin 14-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 20844025-2 2010 This is due to disruption of electron transfer from mutant POR to microsomal cytochrome P450 (CYP) enzymes that play a key role in glucocorticoid and sex steroid synthesis. Steroids 154-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 21067460-6 2010 Desvenlafaxine has clinically insignificant effects on the activity of CYP and P-gp. Desvenlafaxine Succinate 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 20863199-0 2010 Inhibition and induction of human cytochrome P450 enzymes in vitro by capsaicin. Capsaicin 70-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 21062303-6 2010 Other prototypical inhibitors used for characterization of human CYP activities such as furafylline (CYP1A), tranylcypromine (CYP2B) and sulfaphenazole (CYP2C) did not show significant effects in cat and dog liver microsomes. furafylline 88-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 20412007-5 2010 Monooxygenases such as Cytochrome-P450 enzymes do not show any change between the culture systems after 1 day, but exhibit significant up-regulation in CS cultures after 3 days in comparison to hepatocyte monolayers. Cesium 152-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 20863199-2 2010 To investigate potential food-drug or drug-drug interactions, capsaicin was evaluated in vitro against seven human drug-metabolizing cytochrome P450 (CYP) enzymes. Capsaicin 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 20881954-1 2010 Pazopanib, an oral inhibitor of vascular endothelial growth factor receptor, platelet-derived growth factor receptor, and c-kit kinases, inhibits multiple cytochrome P450 (CYP450) enzymes in vitro. pazopanib 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 21226440-11 2010 All patients were prescribed "azole" antifungal agents and five patients were also on high-dose steroids, both agents known to induce cytochrome P-450 enzymes and hence potentiating dapsone toxicity. Steroids 96-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 20945912-1 2010 Menthofuran is a monoterpene present in mint plants that is oxidized by mammalian cytochrome P450 (CYP) to hepatotoxic metabolites. menthofuran 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 20945912-1 2010 Menthofuran is a monoterpene present in mint plants that is oxidized by mammalian cytochrome P450 (CYP) to hepatotoxic metabolites. menthofuran 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-102 20945912-1 2010 Menthofuran is a monoterpene present in mint plants that is oxidized by mammalian cytochrome P450 (CYP) to hepatotoxic metabolites. Monoterpenes 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 20945912-1 2010 Menthofuran is a monoterpene present in mint plants that is oxidized by mammalian cytochrome P450 (CYP) to hepatotoxic metabolites. Monoterpenes 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-102 20881954-1 2010 Pazopanib, an oral inhibitor of vascular endothelial growth factor receptor, platelet-derived growth factor receptor, and c-kit kinases, inhibits multiple cytochrome P450 (CYP450) enzymes in vitro. pazopanib 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-178 20881954-2 2010 This study in patients with advanced cancer evaluated the effect of pazopanib on CYP450 function by comparing the pharmacokinetics of CYP-specific probe drugs in the presence and absence of pazopanib. pazopanib 68-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-87 20881954-2 2010 This study in patients with advanced cancer evaluated the effect of pazopanib on CYP450 function by comparing the pharmacokinetics of CYP-specific probe drugs in the presence and absence of pazopanib. pazopanib 190-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-87 20378604-1 2010 Lipoxygenase, cyclo-oxygenase and cytochrome P450 (CYP) products of arachidonic acid (AA) are implicated in pulmonary vasoregulation. Arachidonic Acid 68-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 20378604-1 2010 Lipoxygenase, cyclo-oxygenase and cytochrome P450 (CYP) products of arachidonic acid (AA) are implicated in pulmonary vasoregulation. Arachidonic Acid 68-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 20378604-2 2010 The CYP-mediated epoxyeicosatrienoates (EETs) have been described previously as the predominant eicosanoids in human lungs upon stimulation with the Ca(2+) ionophore A23187. epoxyeicosatrienoates 17-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 20378604-2 2010 The CYP-mediated epoxyeicosatrienoates (EETs) have been described previously as the predominant eicosanoids in human lungs upon stimulation with the Ca(2+) ionophore A23187. eets 40-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 20378604-2 2010 The CYP-mediated epoxyeicosatrienoates (EETs) have been described previously as the predominant eicosanoids in human lungs upon stimulation with the Ca(2+) ionophore A23187. Eicosanoids 96-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 20378604-2 2010 The CYP-mediated epoxyeicosatrienoates (EETs) have been described previously as the predominant eicosanoids in human lungs upon stimulation with the Ca(2+) ionophore A23187. Calcimycin 166-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 21031626-0 2010 Reversible inhibition of three important human liver cytochrome p450 enzymes by tiliroside. tiliroside 80-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 21071321-8 2010 RESULTS: The development of pseudotumor cerebri after initiation of voriconazole may have been secondary to the inhibition of the cytochrome P450 (CYP) enzymes. Voriconazole 68-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-145 21071321-8 2010 RESULTS: The development of pseudotumor cerebri after initiation of voriconazole may have been secondary to the inhibition of the cytochrome P450 (CYP) enzymes. Voriconazole 68-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 21031626-3 2010 The inhibition of seven CYP isoforms (CYP3A4, CYP1A2, CYP2A6, CYP2D6, CYP2C9, CYP2C8 and CYP2E1) by tiliroside was investigated using in vitro human liver microsomal incubation assays. tiliroside 100-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 21031626-4 2010 The results showed that tiliroside strongly inhibited the activity of CYP3A4 (IC(50) = 9.0 +- 1.7 mum), CYP2C8 (IC(50) = 12.1 +- 0.9 mum) and CYP2C9 (IC(50) = 10.2 +- 0.9 mum) with other CYP isoforms negligibly influenced. tiliroside 24-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 21031626-5 2010 Further kinetic analysis showed that inhibition of these three CYP isoforms by tiliroside is best fit to a competitive way. tiliroside 79-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 21031626-7 2010 The relatively low K(i) values suggested that tiliroside might induce drug-drug interactions with many clinically used drugs which are mainly metabolized by these three CYP isoforms. tiliroside 46-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 20937004-0 2010 In vitro assessment of metabolic drug-drug interaction potential of AZD2624, neurokinin-3 receptor antagonist, through cytochrome P(450) enzyme identification, inhibition, and induction studies. 3-((methylsulfonyl)amino)-2-phenyl-N-(1-phenylpropyl)quinolin-4-carboxamide 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-136 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. 5-sulfoxidation 94-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. 5-sulfoxidation 94-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. mono-n-octyl phthalate 111-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. mono-n-octyl phthalate 111-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. aliphatic- 162-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. aliphatic- 162-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. phenothiazine 177-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. phenothiazine 177-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. Chlorpromazine 203-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 20615392-1 2010 The aim of the present study was to identify cytochrome P450 (CYP) isoenzymes involved in the 5-sulfoxidation, mono-N-demethylation and di-N-demethylation of the aliphatic-type phenothiazine neuroleptic chlorpromazine in human liver. Chlorpromazine 203-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 20615392-3 2010 The obtained results indicate that CYP1A2 is the only CYP isoform that catalyzes the mono-N-demethylation and di-N-demethylation of chlorpromazine (100%) and is the main isoform responsible for chlorpromazine 5-sulfoxidation (64%) at a therapeutic concentration of the drug (10 microM). mono-n-octyl phthalate 85-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 20615392-3 2010 The obtained results indicate that CYP1A2 is the only CYP isoform that catalyzes the mono-N-demethylation and di-N-demethylation of chlorpromazine (100%) and is the main isoform responsible for chlorpromazine 5-sulfoxidation (64%) at a therapeutic concentration of the drug (10 microM). Chlorpromazine 132-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 20615392-3 2010 The obtained results indicate that CYP1A2 is the only CYP isoform that catalyzes the mono-N-demethylation and di-N-demethylation of chlorpromazine (100%) and is the main isoform responsible for chlorpromazine 5-sulfoxidation (64%) at a therapeutic concentration of the drug (10 microM). chlorpromazine 5-sulfoxidation 194-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 20967245-0 2010 Cytochrome P450 reductase: a harbinger of diffusible reduced oxygen species. Oxygen 61-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20967245-1 2010 The bi-enzymatic system of cytochrome P450 (CYP, a hemoprotein) and cytochrome P450 reductase (CPR, a diflavoenzyme) mediate the redox metabolism of diverse indigenous and xenobiotic molecules in various cellular and organ systems, using oxygen and NADPH. Oxygen 238-244 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 20967245-1 2010 The bi-enzymatic system of cytochrome P450 (CYP, a hemoprotein) and cytochrome P450 reductase (CPR, a diflavoenzyme) mediate the redox metabolism of diverse indigenous and xenobiotic molecules in various cellular and organ systems, using oxygen and NADPH. Oxygen 238-244 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 20967245-1 2010 The bi-enzymatic system of cytochrome P450 (CYP, a hemoprotein) and cytochrome P450 reductase (CPR, a diflavoenzyme) mediate the redox metabolism of diverse indigenous and xenobiotic molecules in various cellular and organ systems, using oxygen and NADPH. NADP 249-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 20967245-1 2010 The bi-enzymatic system of cytochrome P450 (CYP, a hemoprotein) and cytochrome P450 reductase (CPR, a diflavoenzyme) mediate the redox metabolism of diverse indigenous and xenobiotic molecules in various cellular and organ systems, using oxygen and NADPH. NADP 249-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 20967245-7 2010 We also quantitatively demonstrate that the rate of oxygen activation and peroxide depletion by CPR accounts for the major reactivity in the CYP+CPR mixture. Oxygen 52-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-144 20967245-7 2010 We also quantitatively demonstrate that the rate of oxygen activation and peroxide depletion by CPR accounts for the major reactivity in the CYP+CPR mixture. Peroxides 74-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-144 20599500-5 2010 Difference between the groups was explained by peculiarities of reductase activity and cytochrome P450 enzyme activities with 7-ethoxyresorufin, 7-methoxyresorufin, 7-methoxycoumarin, 7-benzyloxyresorufin and 7-benzyloxyquinoline. ethoxyresorufin 126-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 20967245-11 2010 Some of the activities that were primarily attributed to the heme-center of CYP are now established to be a facet of the flavins of CPR. Heme 61-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 20967245-11 2010 Some of the activities that were primarily attributed to the heme-center of CYP are now established to be a facet of the flavins of CPR. Flavins 121-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 21603375-11 2010 One of the newer opiates, oxymorphone, has recently been studied as it is metabolized in a non-cytochrome P-450 pathway and therefore bypasses many of the drug-drug interactions common to the elderly. Opiate Alkaloids 17-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 20599500-5 2010 Difference between the groups was explained by peculiarities of reductase activity and cytochrome P450 enzyme activities with 7-ethoxyresorufin, 7-methoxyresorufin, 7-methoxycoumarin, 7-benzyloxyresorufin and 7-benzyloxyquinoline. 7-methoxyresorufin 145-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 20599500-5 2010 Difference between the groups was explained by peculiarities of reductase activity and cytochrome P450 enzyme activities with 7-ethoxyresorufin, 7-methoxyresorufin, 7-methoxycoumarin, 7-benzyloxyresorufin and 7-benzyloxyquinoline. herniarin 165-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 20599500-5 2010 Difference between the groups was explained by peculiarities of reductase activity and cytochrome P450 enzyme activities with 7-ethoxyresorufin, 7-methoxyresorufin, 7-methoxycoumarin, 7-benzyloxyresorufin and 7-benzyloxyquinoline. benzyloxyresorufin 184-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 20599500-5 2010 Difference between the groups was explained by peculiarities of reductase activity and cytochrome P450 enzyme activities with 7-ethoxyresorufin, 7-methoxyresorufin, 7-methoxycoumarin, 7-benzyloxyresorufin and 7-benzyloxyquinoline. 7-benzyloxyquinoline 209-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 20634336-6 2010 Incubations with recombinant flavin-containing monooxygenase (FMO) and UDP-glucuronosyltransferase (UGT) enzymes and inhibition studies using the nonselective cytochrome P450 (P450) chemical inhibitor 1-aminobenzotriazole in human hepatocytes indicated that FMO1 and FMO3 contributed to cediranib N-oxidation, whereas UGT1A4 had a major role in cediranib N(+)-glucuronidation. 1H-1,2,3-benzotriazol-4-amine 203-221 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-202 21603375-11 2010 One of the newer opiates, oxymorphone, has recently been studied as it is metabolized in a non-cytochrome P-450 pathway and therefore bypasses many of the drug-drug interactions common to the elderly. Oxymorphone 26-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 20877451-5 2010 Cytochrome P450 (CYP) enzymes, CYP2D6 in particular, play a major role in the metabolism of tamoxifen to active metabolites. Tamoxifen 92-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20399842-2 2010 BP is converted in liver and lung to benzo(a)pyrene-7,8-diol-9,10-epoxide (BPDE) by the enzymes of the cytochrome P450 (CYP) superfamily, namely CYP1A1/1A2, and CYP1B1. Benzo(a)pyrene 0-2 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 20399842-2 2010 BP is converted in liver and lung to benzo(a)pyrene-7,8-diol-9,10-epoxide (BPDE) by the enzymes of the cytochrome P450 (CYP) superfamily, namely CYP1A1/1A2, and CYP1B1. Benzo(a)pyrene 0-2 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 20399842-2 2010 BP is converted in liver and lung to benzo(a)pyrene-7,8-diol-9,10-epoxide (BPDE) by the enzymes of the cytochrome P450 (CYP) superfamily, namely CYP1A1/1A2, and CYP1B1. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 37-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 20399842-2 2010 BP is converted in liver and lung to benzo(a)pyrene-7,8-diol-9,10-epoxide (BPDE) by the enzymes of the cytochrome P450 (CYP) superfamily, namely CYP1A1/1A2, and CYP1B1. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 37-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 20399842-2 2010 BP is converted in liver and lung to benzo(a)pyrene-7,8-diol-9,10-epoxide (BPDE) by the enzymes of the cytochrome P450 (CYP) superfamily, namely CYP1A1/1A2, and CYP1B1. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 75-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 20399842-2 2010 BP is converted in liver and lung to benzo(a)pyrene-7,8-diol-9,10-epoxide (BPDE) by the enzymes of the cytochrome P450 (CYP) superfamily, namely CYP1A1/1A2, and CYP1B1. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 75-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 20877451-5 2010 Cytochrome P450 (CYP) enzymes, CYP2D6 in particular, play a major role in the metabolism of tamoxifen to active metabolites. Tamoxifen 92-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 20652238-1 2010 The angular dioxygenase, cytochrome P450, lignin peroxidase, and dehalogenase are known as dioxin-metabolizing enzymes. Dioxins 91-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-59 20735423-1 2010 BACKGROUND AND PURPOSE: The conversion of clopidogrel to its active metabolite, R-130964, is a two-step cytochrome P450 (CYP)-dependent process. Clopidogrel 42-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 20735423-1 2010 BACKGROUND AND PURPOSE: The conversion of clopidogrel to its active metabolite, R-130964, is a two-step cytochrome P450 (CYP)-dependent process. Clopidogrel 42-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 20735423-2 2010 The current investigations were performed to characterize in vitro the effects of different CYP inhibitors on the biotransformation and on the antiplatelet effect of clopidogrel. Clopidogrel 166-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 24278521-0 2010 Bioactivation of Aromatic Amines by Human CYP2W1, An Orphan Cytochrome P450 Enzyme. aromatic amines 17-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 20629947-0 2010 Measurement and diagnostic use of hepatic cytochrome P450 metabolism of oleic acid in liver disease. Oleic Acid 72-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 20629947-2 2010 As of today, the contribution of individual cytochrome P450 (CYP) mono-oxygenases to the epoxidation of this fatty acid is unknown. Fatty Acids 109-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 20629947-2 2010 As of today, the contribution of individual cytochrome P450 (CYP) mono-oxygenases to the epoxidation of this fatty acid is unknown. Fatty Acids 109-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 20629947-5 2010 Fifteen CYP mono-oxygenases were investigated in vitro as a potential source of cis-EODA. 9,10-epoxystearic acid 80-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-11 20629947-7 2010 Production of cis-EODA was catalysed by CYP in the following order: 2C8, 2C9, 2C19, 3A4, 1A2 and CYP3A7. 9,10-epoxystearic acid 14-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 20699675-5 2010 Because pitavastatin is minimally metabolized by the cytochrome P-450 isoenzymes, it is associated with a lower frequency of drug-drug interactions, and this may be a desirable characteristic of pitavastatin. pitavastatin 8-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 20699675-5 2010 Because pitavastatin is minimally metabolized by the cytochrome P-450 isoenzymes, it is associated with a lower frequency of drug-drug interactions, and this may be a desirable characteristic of pitavastatin. pitavastatin 195-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 20493836-1 2010 Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid (AA) catalyzed by cytochrome P450 (CYP), have many essential biologic roles in the cardiovascular system including inhibition of apoptosis in cardiomyocytes. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 20493836-1 2010 Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid (AA) catalyzed by cytochrome P450 (CYP), have many essential biologic roles in the cardiovascular system including inhibition of apoptosis in cardiomyocytes. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 19904716-2 2010 This method was established by the characteristics of recombinant cytochrome P-450 (CYP) isozymes, where CYP2C19 prefers to metabolize R-methadone and CYP2B6 prefers to metabolize S-methadone. Methadone 135-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 20493836-1 2010 Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid (AA) catalyzed by cytochrome P450 (CYP), have many essential biologic roles in the cardiovascular system including inhibition of apoptosis in cardiomyocytes. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 20493836-1 2010 Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid (AA) catalyzed by cytochrome P450 (CYP), have many essential biologic roles in the cardiovascular system including inhibition of apoptosis in cardiomyocytes. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 20493836-1 2010 Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid (AA) catalyzed by cytochrome P450 (CYP), have many essential biologic roles in the cardiovascular system including inhibition of apoptosis in cardiomyocytes. Arachidonic Acid 49-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 20493836-1 2010 Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid (AA) catalyzed by cytochrome P450 (CYP), have many essential biologic roles in the cardiovascular system including inhibition of apoptosis in cardiomyocytes. Arachidonic Acid 49-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 20532872-7 2010 Polymorphism in the metabolic enzyme cytochrome P-450 is associated with risk of myocardial infarction in caffeine users. Caffeine 106-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 19804952-3 2010 The goal of this study is to investigate the oxidative metabolites of SA formed during incubations with rat liver microsomes (RLM) and recombinant human cytochrome P450 (CYP) and to tentatively identify the CYP isoforms involved in SA detoxification. sanguinarine 70-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-168 19804952-3 2010 The goal of this study is to investigate the oxidative metabolites of SA formed during incubations with rat liver microsomes (RLM) and recombinant human cytochrome P450 (CYP) and to tentatively identify the CYP isoforms involved in SA detoxification. sanguinarine 70-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-173 19804952-3 2010 The goal of this study is to investigate the oxidative metabolites of SA formed during incubations with rat liver microsomes (RLM) and recombinant human cytochrome P450 (CYP) and to tentatively identify the CYP isoforms involved in SA detoxification. sanguinarine 70-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-210 19804952-3 2010 The goal of this study is to investigate the oxidative metabolites of SA formed during incubations with rat liver microsomes (RLM) and recombinant human cytochrome P450 (CYP) and to tentatively identify the CYP isoforms involved in SA detoxification. sanguinarine 232-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-210 19804952-6 2010 The main metabolite M2 (m/z 320) resulted from ring-cleavage of SA followed by demethylation, whereas M4 (m/z 348) is oxidized by CYP in the presence of NADPH. NADP 153-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 20610983-6 2010 Although new triazole antifungals are well tolerated, they still cause significant inhibition of CYP enzymes. Triazoles 13-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 20610983-8 2010 Azole antimycotics are amongst the strongest inhibitors of CYP-mediated drug metabolism. Azoles 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 20658791-7 2010 Duloxetine and escitalopram are extensively metabolized via cytochrome P450 (CYP) enzymes and the decreased hepatic metabolism present in many older adults should be taken into account when prescribing these medications. Duloxetine Hydrochloride 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 20658791-7 2010 Duloxetine and escitalopram are extensively metabolized via cytochrome P450 (CYP) enzymes and the decreased hepatic metabolism present in many older adults should be taken into account when prescribing these medications. Duloxetine Hydrochloride 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 20658791-7 2010 Duloxetine and escitalopram are extensively metabolized via cytochrome P450 (CYP) enzymes and the decreased hepatic metabolism present in many older adults should be taken into account when prescribing these medications. Citalopram 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 20658791-7 2010 Duloxetine and escitalopram are extensively metabolized via cytochrome P450 (CYP) enzymes and the decreased hepatic metabolism present in many older adults should be taken into account when prescribing these medications. Citalopram 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 19904716-2 2010 This method was established by the characteristics of recombinant cytochrome P-450 (CYP) isozymes, where CYP2C19 prefers to metabolize R-methadone and CYP2B6 prefers to metabolize S-methadone. Methadone 135-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 19904716-2 2010 This method was established by the characteristics of recombinant cytochrome P-450 (CYP) isozymes, where CYP2C19 prefers to metabolize R-methadone and CYP2B6 prefers to metabolize S-methadone. Methadone 180-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 19904716-2 2010 This method was established by the characteristics of recombinant cytochrome P-450 (CYP) isozymes, where CYP2C19 prefers to metabolize R-methadone and CYP2B6 prefers to metabolize S-methadone. Methadone 180-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 20052538-2 2010 Orally administered tamoxifen, is extensively metabolized by cytochrome P450 (CYP) enzymes, namely CYP3A4 and CYP2D6, into active metabolites, especially endoxifen. Tamoxifen 20-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 20382753-0 2010 Cytochrome P450-mediated bioactivation of the epidermal growth factor receptor inhibitor erlotinib to a reactive electrophile. Erlotinib Hydrochloride 89-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20382753-5 2010 Cytochrome P450 (P450)-dependent adducts are proposed to be formed via reactive epoxide and electrophilic quinone-imine intermediates. reactive epoxide 71-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20382753-5 2010 Cytochrome P450 (P450)-dependent adducts are proposed to be formed via reactive epoxide and electrophilic quinone-imine intermediates. quinone-imine 106-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 20052538-2 2010 Orally administered tamoxifen, is extensively metabolized by cytochrome P450 (CYP) enzymes, namely CYP3A4 and CYP2D6, into active metabolites, especially endoxifen. Tamoxifen 20-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 20052538-2 2010 Orally administered tamoxifen, is extensively metabolized by cytochrome P450 (CYP) enzymes, namely CYP3A4 and CYP2D6, into active metabolites, especially endoxifen. 4-hydroxy-N-desmethyltamoxifen 154-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 20052538-2 2010 Orally administered tamoxifen, is extensively metabolized by cytochrome P450 (CYP) enzymes, namely CYP3A4 and CYP2D6, into active metabolites, especially endoxifen. 4-hydroxy-N-desmethyltamoxifen 154-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 20570348-2 2010 The heterocyclic amine, 2-amino-3-methylimidazo[4,5-f]quinoline (IQ) is a food-borne carcinogen being metabolically activated by cytochrome P450 (CYP) enzymes, especially by CYP1A1/2. heterocyclic amine 4-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-144 20570348-2 2010 The heterocyclic amine, 2-amino-3-methylimidazo[4,5-f]quinoline (IQ) is a food-borne carcinogen being metabolically activated by cytochrome P450 (CYP) enzymes, especially by CYP1A1/2. heterocyclic amine 4-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 20570348-2 2010 The heterocyclic amine, 2-amino-3-methylimidazo[4,5-f]quinoline (IQ) is a food-borne carcinogen being metabolically activated by cytochrome P450 (CYP) enzymes, especially by CYP1A1/2. 2-amino-3-methylimidazo(4,5-f)quinoline 24-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-144 20570348-2 2010 The heterocyclic amine, 2-amino-3-methylimidazo[4,5-f]quinoline (IQ) is a food-borne carcinogen being metabolically activated by cytochrome P450 (CYP) enzymes, especially by CYP1A1/2. 2-amino-3-methylimidazo(4,5-f)quinoline 24-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 20408551-4 2010 Unwanted side effects are preventable without remarkable loss of activity when the responsible constituent(s) are carefully removed during the extraction process, as demonstrated for hyperforin (3), which is responsible for the induction of cytochrome P450 (CYP)-metabolizing enzymes (CYP3A4, in particular). hyperforin 183-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-256 20606811-2 2010 The cytochrome P(450) isoform (CYP2E1) specifically involved in ethanol oxidation is discussed. Ethanol 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-21 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. Carbon 17-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. -hydroxyethyl 35-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. Oxygen 54-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. centered (hydroxyl) 61-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. Ethanol 115-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. hydroxyethyl radicals 156-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. Oxygen 244-252 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. (p(450)-fe(3+)o 328-343 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. Heme 384-388 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20606811-6 2010 The formation of carbon-centered (1-hydroxyethyl) and oxygen-centered (hydroxyl) radicals during the metabolism of ethanol is considered: the generation of hydroxyethyl radicals, which occurs likely during the process of univalent reduction of dioxygen, is highlighted and is carried out by ferric cytochrome P(450) oxy-complex (P(450)-Fe(3+)O(2) (.-)) formed during the reduction of heme-oxygen. Oxygen 246-252 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 298-315 20408551-4 2010 Unwanted side effects are preventable without remarkable loss of activity when the responsible constituent(s) are carefully removed during the extraction process, as demonstrated for hyperforin (3), which is responsible for the induction of cytochrome P450 (CYP)-metabolizing enzymes (CYP3A4, in particular). hyperforin 183-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-261 19513837-8 2010 This leak may pose a clinical danger by increasing drug uptake into the bloodstream, a phenomenon which would act synergistically with the effect of omeprazole on inhibiting liver cytochrome P450s that remove drugs from the bloodstream, thereby elevating drug blood levels. Omeprazole 149-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-195 20437462-0 2010 Contributions of human cytochrome P450 enzymes to glyburide metabolism. Glyburide 50-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 20437462-3 2010 Therefore, there has been a continued interest in identifying human cytochrome P450 (CYP) isoforms that play a major role in the metabolism of GLB. Glyburide 143-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 20437462-3 2010 Therefore, there has been a continued interest in identifying human cytochrome P450 (CYP) isoforms that play a major role in the metabolism of GLB. Glyburide 143-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 20437462-5 2010 The present study systematically investigated the contributions of various human CYP isoforms (CYP3A4, CYP3A5, CYP2C8, CYP2C9 and CYP2C19) to in vitro metabolism of GLB. Glyburide 165-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 20437462-6 2010 GLB depletion and metabolite formation in human liver microsomes were most significantly inhibited by the CYP3A inhibitor ketoconazole compared with the inhibitors of other CYP isoforms. Glyburide 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 20437462-6 2010 GLB depletion and metabolite formation in human liver microsomes were most significantly inhibited by the CYP3A inhibitor ketoconazole compared with the inhibitors of other CYP isoforms. Ketoconazole 122-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 20437462-7 2010 Furthermore, multiple correlation analysis between GLB depletion and individual CYP activities was performed, demonstrating a significant correlation between GLB depletion and the CYP3A probe activity in 16 individual human liver microsomal preparations, but not between GLB depletion and the CYP2C19, CYP2C8 or CYP2C9 probe activity. Glyburide 158-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 20437462-7 2010 Furthermore, multiple correlation analysis between GLB depletion and individual CYP activities was performed, demonstrating a significant correlation between GLB depletion and the CYP3A probe activity in 16 individual human liver microsomal preparations, but not between GLB depletion and the CYP2C19, CYP2C8 or CYP2C9 probe activity. Glyburide 158-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 20437462-8 2010 By using recombinant supersomes overexpressing individual human CYP isoforms, it was found that GLB could be depleted by all the enzymes tested; however, the intrinsic clearance (V(max)/K(m)) of CYP3A4 for GLB depletion was 4-17 times greater than that of other CYP isoforms. Glyburide 96-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 20437462-8 2010 By using recombinant supersomes overexpressing individual human CYP isoforms, it was found that GLB could be depleted by all the enzymes tested; however, the intrinsic clearance (V(max)/K(m)) of CYP3A4 for GLB depletion was 4-17 times greater than that of other CYP isoforms. Glyburide 96-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-198 20437462-8 2010 By using recombinant supersomes overexpressing individual human CYP isoforms, it was found that GLB could be depleted by all the enzymes tested; however, the intrinsic clearance (V(max)/K(m)) of CYP3A4 for GLB depletion was 4-17 times greater than that of other CYP isoforms. Glyburide 206-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 20437462-8 2010 By using recombinant supersomes overexpressing individual human CYP isoforms, it was found that GLB could be depleted by all the enzymes tested; however, the intrinsic clearance (V(max)/K(m)) of CYP3A4 for GLB depletion was 4-17 times greater than that of other CYP isoforms. Glyburide 206-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-198 20124396-7 2010 Lersivirine was extensively metabolized, primarily by UDP glucuronosyltransferase- and cytochrome P450-dependent pathways, with 22 metabolites being identified in this study. UK 453,061 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 20609060-4 2010 KEY FINDINGS: The results showed that nateglinide inhibited CYP2C9 and CYP2C19 with an IC50(app) (apparent value of the 50% inhibitory concentration) of 125 micromol/l and 946 micromol/l, respectively, while M1 did not inhibit any of the CYP isoforms. Nateglinide 38-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 20609060-0 2010 Inhibition of human liver microsomal CYP by nateglinide. Nateglinide 44-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 20609060-8 2010 The results revealed that the inhibition of CYP by nateglinide was not influenced by pre-incubation, and that the possibility of MBI is very low. Nateglinide 51-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 20609060-1 2010 OBJECTIVES: Nateglinide is metabolized by CYP2C9 and CYP3A4, therefore drug-drug interactions through cytochrome P450 (CYP) inhibition may occur. Nateglinide 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-117 20609060-9 2010 CONCLUSIONS: The possibility of drug-drug interactions involving nateglinide that might be attributable to CYP inhibition is low. Nateglinide 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-110 20609060-1 2010 OBJECTIVES: Nateglinide is metabolized by CYP2C9 and CYP3A4, therefore drug-drug interactions through cytochrome P450 (CYP) inhibition may occur. Nateglinide 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 20218941-6 2010 In addition to CYP gene expression, nuclear receptor proteins regulate the expression of complex gene networks, and therefore mediate the metabolism and modify the effects of steroid hormones, fat-soluble vitamins, and free fatty acids on the metabolic, reproductive, and developmental processes of mammals. Steroids 175-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 20609060-2 2010 In this study, we examined the inhibitory effects of nateglinide and its major metabolite N-[trans-4-(1-hydroxy-1-methylethyl)-cyclohexanecarbonyl]-D-phenylalanine (M1) on various CYP isoforms in human liver microsomes. n-[trans-4-(1-hydroxy-1-methylethyl)-cyclohexanecarbonyl]-d-phenylalanine 90-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 20218941-6 2010 In addition to CYP gene expression, nuclear receptor proteins regulate the expression of complex gene networks, and therefore mediate the metabolism and modify the effects of steroid hormones, fat-soluble vitamins, and free fatty acids on the metabolic, reproductive, and developmental processes of mammals. Fatty Acids, Nonesterified 219-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 19998329-0 2010 Cytochrome P450-dependent toxicity of dapsone in human erythrocytes. Dapsone 38-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 19998329-2 2010 An in vitro microsomal/cytochrome P(450) (CYP)-linked assay, which allows reactive metabolites generated in situ to react with the co-incubated human erythrocytes, was employed to profile CYP isoforms responsible for hemotoxicity of dapsone. Dapsone 233-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-40 20207119-1 2010 Polyunsaturated fatty acids of nutritional value may affect cell functions after their release from cell lipid storage sites, especially phospholipids, and specific oxygenation by cyclooxygenases, lipoxygenases and cytochrome P(450). Fatty Acids, Unsaturated 0-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-232 19562511-1 2010 Acenocoumarol is mainly catabolized by CYP2C9 isoform of cytochrome P450 (CYP) liver complex and exerts its anticoagulant effect through the inhibition of Vitamin K Epoxide Reductase (VKOR). Acenocoumarol 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 19562511-1 2010 Acenocoumarol is mainly catabolized by CYP2C9 isoform of cytochrome P450 (CYP) liver complex and exerts its anticoagulant effect through the inhibition of Vitamin K Epoxide Reductase (VKOR). Acenocoumarol 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 20051482-1 2010 Cytochrome P450 (P450)1A1 plays a critical role in the metabolic activation and detoxification of polycyclic aromatic hydrocarbons (PAHs), many of which are potent human carcinogens. Polycyclic Aromatic Hydrocarbons 98-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-25 20051482-1 2010 Cytochrome P450 (P450)1A1 plays a critical role in the metabolic activation and detoxification of polycyclic aromatic hydrocarbons (PAHs), many of which are potent human carcinogens. Polycyclic Aromatic Hydrocarbons 132-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-25 20350136-1 2010 AIMS: The antidepressant escitalopram (S-CIT) is metabolized by the cytochrome-P450 (CYP) enzymes CYP 2D6, 2C19 and 3A4. Citalopram 25-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 20350136-1 2010 AIMS: The antidepressant escitalopram (S-CIT) is metabolized by the cytochrome-P450 (CYP) enzymes CYP 2D6, 2C19 and 3A4. Citalopram 25-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 20350136-1 2010 AIMS: The antidepressant escitalopram (S-CIT) is metabolized by the cytochrome-P450 (CYP) enzymes CYP 2D6, 2C19 and 3A4. Sulfur 3-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 20350136-1 2010 AIMS: The antidepressant escitalopram (S-CIT) is metabolized by the cytochrome-P450 (CYP) enzymes CYP 2D6, 2C19 and 3A4. Sulfur 3-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 20218935-0 2010 Rapid screening of commercially available herbal products for the inhibition of major human hepatic cytochrome P450 enzymes using the N-in-one cocktail. n-in-one 134-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 20061389-1 2010 Cytochrome P450 (P450) 2A6 activates nitrosamines, including N,N-dimethylnitrosamine (DMN) and N,N-diethylnitrosamine (DEN), to alkyl diazohydroxides (which are DNA-alkylating agents) and also aldehydes (HCHO from DMN and CH(3)CHO from DEN). Nitrosamines 37-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 20153303-7 2010 As a basis for additionally coordinating and focusing research on critical data gaps in a risk assessment context, five key events in the mode of action for benzene-induced leukemia are proposed: (1) benzene metabolism via Cytochrome P450, (2) the interaction of benzene metabolites with target cells in the bone marrow, (3) formation of initiated, mutated target cells, (4) selective proliferation of the mutated cells and (5) production of leukemia. Benzene 157-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-238 20153303-7 2010 As a basis for additionally coordinating and focusing research on critical data gaps in a risk assessment context, five key events in the mode of action for benzene-induced leukemia are proposed: (1) benzene metabolism via Cytochrome P450, (2) the interaction of benzene metabolites with target cells in the bone marrow, (3) formation of initiated, mutated target cells, (4) selective proliferation of the mutated cells and (5) production of leukemia. Benzene 200-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-238 20153303-7 2010 As a basis for additionally coordinating and focusing research on critical data gaps in a risk assessment context, five key events in the mode of action for benzene-induced leukemia are proposed: (1) benzene metabolism via Cytochrome P450, (2) the interaction of benzene metabolites with target cells in the bone marrow, (3) formation of initiated, mutated target cells, (4) selective proliferation of the mutated cells and (5) production of leukemia. Benzene 200-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-238 20036648-5 2010 CYP4F3A, which encodes the leukotriene B(4) (LTB(4)) omega-hydroxylase, is important for inactivation of LTB(4) in neutrophils. Leukotriene B4 27-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-70 20061389-1 2010 Cytochrome P450 (P450) 2A6 activates nitrosamines, including N,N-dimethylnitrosamine (DMN) and N,N-diethylnitrosamine (DEN), to alkyl diazohydroxides (which are DNA-alkylating agents) and also aldehydes (HCHO from DMN and CH(3)CHO from DEN). Dimethylnitrosamine 61-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 20061389-1 2010 Cytochrome P450 (P450) 2A6 activates nitrosamines, including N,N-dimethylnitrosamine (DMN) and N,N-diethylnitrosamine (DEN), to alkyl diazohydroxides (which are DNA-alkylating agents) and also aldehydes (HCHO from DMN and CH(3)CHO from DEN). Dimethylnitrosamine 86-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 20061389-1 2010 Cytochrome P450 (P450) 2A6 activates nitrosamines, including N,N-dimethylnitrosamine (DMN) and N,N-diethylnitrosamine (DEN), to alkyl diazohydroxides (which are DNA-alkylating agents) and also aldehydes (HCHO from DMN and CH(3)CHO from DEN). Diethylnitrosamine 95-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 20061389-1 2010 Cytochrome P450 (P450) 2A6 activates nitrosamines, including N,N-dimethylnitrosamine (DMN) and N,N-diethylnitrosamine (DEN), to alkyl diazohydroxides (which are DNA-alkylating agents) and also aldehydes (HCHO from DMN and CH(3)CHO from DEN). Diethylnitrosamine 119-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 20061389-1 2010 Cytochrome P450 (P450) 2A6 activates nitrosamines, including N,N-dimethylnitrosamine (DMN) and N,N-diethylnitrosamine (DEN), to alkyl diazohydroxides (which are DNA-alkylating agents) and also aldehydes (HCHO from DMN and CH(3)CHO from DEN). alkyl diazohydroxides 128-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 20061389-1 2010 Cytochrome P450 (P450) 2A6 activates nitrosamines, including N,N-dimethylnitrosamine (DMN) and N,N-diethylnitrosamine (DEN), to alkyl diazohydroxides (which are DNA-alkylating agents) and also aldehydes (HCHO from DMN and CH(3)CHO from DEN). Aldehydes 193-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 20061389-1 2010 Cytochrome P450 (P450) 2A6 activates nitrosamines, including N,N-dimethylnitrosamine (DMN) and N,N-diethylnitrosamine (DEN), to alkyl diazohydroxides (which are DNA-alkylating agents) and also aldehydes (HCHO from DMN and CH(3)CHO from DEN). Dimethylnitrosamine 214-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 20061389-1 2010 Cytochrome P450 (P450) 2A6 activates nitrosamines, including N,N-dimethylnitrosamine (DMN) and N,N-diethylnitrosamine (DEN), to alkyl diazohydroxides (which are DNA-alkylating agents) and also aldehydes (HCHO from DMN and CH(3)CHO from DEN). Diethylnitrosamine 236-239 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 20393001-5 2010 An ex vivo model of cryopreserved human hepatocytes was used to evaluate the CYP450 metabolism induction potential of curcumin for CYP P450 3A4, 2C8/2C9, and 2D6. Curcumin 118-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-83 20393001-7 2010 At a curcumin concentration of 58.3 microM, 10.5% and 22.5% inhibition of CYP450 2C9 and CYP450 2C8 activity, respectively, was observed. Curcumin 5-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-80 20393001-7 2010 At a curcumin concentration of 58.3 microM, 10.5% and 22.5% inhibition of CYP450 2C9 and CYP450 2C8 activity, respectively, was observed. Curcumin 5-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-95 20393001-10 2010 CONCLUSION: There is low potential for CYP450 mediated drug interactions at physiologic serum concentrations of liposomal curcumin. Curcumin 122-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-45 20393001-11 2010 Based on preliminary data, liposomal curcumin will not interact with other chemotherapy agents that are metabolized and/or eliminated via the primary drug metabolizing CYP450 pathways. Curcumin 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-174 20002521-3 2010 Rasagiline is primarily metabolized by hepatic cytochrome P-450 to form its major metabolite, 1-(R)-aminoindan, a non-amphetamine, weak reversible MAO-B inhibitor compound. (R)-(-)-1-Aminoindan 94-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 20002521-3 2010 Rasagiline is primarily metabolized by hepatic cytochrome P-450 to form its major metabolite, 1-(R)-aminoindan, a non-amphetamine, weak reversible MAO-B inhibitor compound. rasagiline 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 20002521-3 2010 Rasagiline is primarily metabolized by hepatic cytochrome P-450 to form its major metabolite, 1-(R)-aminoindan, a non-amphetamine, weak reversible MAO-B inhibitor compound. Amphetamine 118-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 20180610-2 2010 However, clopidogrel is a prodrug that needs to be metabolized to the active thiol metabolite by the cytochrome P450 (CYP) system. Clopidogrel 9-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 20180610-2 2010 However, clopidogrel is a prodrug that needs to be metabolized to the active thiol metabolite by the cytochrome P450 (CYP) system. Clopidogrel 9-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 20180610-2 2010 However, clopidogrel is a prodrug that needs to be metabolized to the active thiol metabolite by the cytochrome P450 (CYP) system. Sulfhydryl Compounds 77-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 20180610-2 2010 However, clopidogrel is a prodrug that needs to be metabolized to the active thiol metabolite by the cytochrome P450 (CYP) system. Sulfhydryl Compounds 77-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). Arachidonic Acid 90-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 19931366-5 2010 The aromatic hydroxylation of alpha-ZAL is almost exclusively mediated by the human cytochrome P450 (hCYP) 1A2 isoform. Zeranol 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-110 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). Arachidonic Acid 90-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). epoxyeicosatrienoic acids 110-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). epoxyeicosatrienoic acids 110-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). eets 137-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). eets 137-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). dihydroxyeicosatrienoic acids 176-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). dihydroxyeicosatrienoic acids 176-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). dhets 207-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). dhets 207-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 19904241-2 2010 Clopidogrel is converted to its active metabolite by cytochrome P450 (CYP) enzymes. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 19919843-0 2010 Impact of genetic and acquired alteration in cytochrome P450 system on pharmacologic and clinical response to clopidogrel. Clopidogrel 110-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 19919843-2 2010 Clopidogrel, a thienopyridine, is a prodrug that is transformed in vivo to an active metabolite by the cytochrome P450 (CYP) enzyme system. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 19919843-2 2010 Clopidogrel, a thienopyridine, is a prodrug that is transformed in vivo to an active metabolite by the cytochrome P450 (CYP) enzyme system. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 19919843-2 2010 Clopidogrel, a thienopyridine, is a prodrug that is transformed in vivo to an active metabolite by the cytochrome P450 (CYP) enzyme system. thienopyridine 15-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 19919843-2 2010 Clopidogrel, a thienopyridine, is a prodrug that is transformed in vivo to an active metabolite by the cytochrome P450 (CYP) enzyme system. thienopyridine 15-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 19919843-4 2010 Recent data demonstrated patients carrying a genetic variant of CYP enzymes (e.g. CYP2C19) would have a higher rate of ischemic events than non-carriers due to an attenuation of the pharmacokinetic and pharmacodynamic responses to clopidogrel. Clopidogrel 231-242 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 19919843-6 2010 PPIs are extensively metabolized by the cytochrome P450 system and have been associated with decreased antiplatelet activity of clopidogrel. Clopidogrel 128-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 19904241-2 2010 Clopidogrel is converted to its active metabolite by cytochrome P450 (CYP) enzymes. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 19904241-12 2010 As the presence of PPIs and clopidogrel in plasma is short lived, separation by 12-20 h should in theory prevent competitive inhibition of CYP metabolism and minimize any potential, though unproven, clinical interaction. Clopidogrel 28-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 20001672-2 2010 The present study aimed to profile the phase I metabolites of schizandrin and identify the cytochrome P450 (CYP) isoforms involved. schizandrin 62-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 20923245-2 2010 Ambrisentan is primarily metabolized by glucuronidation and undergoes cytochrome P450 (CYP)-mediated oxidation to a lesser extent. ambrisentan 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 20923245-2 2010 Ambrisentan is primarily metabolized by glucuronidation and undergoes cytochrome P450 (CYP)-mediated oxidation to a lesser extent. ambrisentan 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 20425602-2 2010 Biotransformation of tamoxifen to the potent antiestrogen endoxifen is performed by cytochrome P450 (CYP) enzymes, in particular the CYP2D6 isoform. Tamoxifen 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 20425602-2 2010 Biotransformation of tamoxifen to the potent antiestrogen endoxifen is performed by cytochrome P450 (CYP) enzymes, in particular the CYP2D6 isoform. Tamoxifen 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 20425602-2 2010 Biotransformation of tamoxifen to the potent antiestrogen endoxifen is performed by cytochrome P450 (CYP) enzymes, in particular the CYP2D6 isoform. 4-hydroxy-N-desmethyltamoxifen 58-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 20425602-2 2010 Biotransformation of tamoxifen to the potent antiestrogen endoxifen is performed by cytochrome P450 (CYP) enzymes, in particular the CYP2D6 isoform. 4-hydroxy-N-desmethyltamoxifen 58-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 20002088-4 2009 * This cocktail can be used to test the effects of a new chemical entity on multiple CYP isoforms in a single clinical study: CYP1A2 (caffeine), CYP2C9 (warfarin), CYP2C19 (omeprazole), CYP2D6 (metoprolol), and CYP3A (midazolam) and was designed to overcome potential liabilities of other reported cocktails. Caffeine 134-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 19615687-1 2009 BACKGROUND: Cytochrome P450 (CYP) is expressed in the human endothelium and metabolizes arachidonic acid into vasoactive epoxyeicosatrienoic and 20-hydroxyeicosatetraenoic acids. Arachidonic Acid 88-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 19615687-1 2009 BACKGROUND: Cytochrome P450 (CYP) is expressed in the human endothelium and metabolizes arachidonic acid into vasoactive epoxyeicosatrienoic and 20-hydroxyeicosatetraenoic acids. Arachidonic Acid 88-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 19615687-1 2009 BACKGROUND: Cytochrome P450 (CYP) is expressed in the human endothelium and metabolizes arachidonic acid into vasoactive epoxyeicosatrienoic and 20-hydroxyeicosatetraenoic acids. epoxyeicosatrienoic 121-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 19615687-1 2009 BACKGROUND: Cytochrome P450 (CYP) is expressed in the human endothelium and metabolizes arachidonic acid into vasoactive epoxyeicosatrienoic and 20-hydroxyeicosatetraenoic acids. epoxyeicosatrienoic 121-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 19615687-1 2009 BACKGROUND: Cytochrome P450 (CYP) is expressed in the human endothelium and metabolizes arachidonic acid into vasoactive epoxyeicosatrienoic and 20-hydroxyeicosatetraenoic acids. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 145-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 19615687-1 2009 BACKGROUND: Cytochrome P450 (CYP) is expressed in the human endothelium and metabolizes arachidonic acid into vasoactive epoxyeicosatrienoic and 20-hydroxyeicosatetraenoic acids. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 145-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 19615687-2 2009 CYP enzymes have been linked to hypertension and generation of reactive oxygen species. Reactive Oxygen Species 63-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 19932643-1 2009 A rapid simultaneous determination method for in vitro Cytochrome P450 (CYP) activity assay of 1,2,3,4-tetrahydroacridin-9-amine (tacrine) metabolites using ultra high performance liquid chromatography (UHPLC) coupled with computer-assisted in-source collision induced dissociation (CID) monitoring was investigated. Tacrine 95-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 19932643-1 2009 A rapid simultaneous determination method for in vitro Cytochrome P450 (CYP) activity assay of 1,2,3,4-tetrahydroacridin-9-amine (tacrine) metabolites using ultra high performance liquid chromatography (UHPLC) coupled with computer-assisted in-source collision induced dissociation (CID) monitoring was investigated. Tacrine 95-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 19932643-1 2009 A rapid simultaneous determination method for in vitro Cytochrome P450 (CYP) activity assay of 1,2,3,4-tetrahydroacridin-9-amine (tacrine) metabolites using ultra high performance liquid chromatography (UHPLC) coupled with computer-assisted in-source collision induced dissociation (CID) monitoring was investigated. Tacrine 130-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 19932643-1 2009 A rapid simultaneous determination method for in vitro Cytochrome P450 (CYP) activity assay of 1,2,3,4-tetrahydroacridin-9-amine (tacrine) metabolites using ultra high performance liquid chromatography (UHPLC) coupled with computer-assisted in-source collision induced dissociation (CID) monitoring was investigated. Tacrine 130-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 20002088-4 2009 * This cocktail can be used to test the effects of a new chemical entity on multiple CYP isoforms in a single clinical study: CYP1A2 (caffeine), CYP2C9 (warfarin), CYP2C19 (omeprazole), CYP2D6 (metoprolol), and CYP3A (midazolam) and was designed to overcome potential liabilities of other reported cocktails. Warfarin 153-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 20002088-4 2009 * This cocktail can be used to test the effects of a new chemical entity on multiple CYP isoforms in a single clinical study: CYP1A2 (caffeine), CYP2C9 (warfarin), CYP2C19 (omeprazole), CYP2D6 (metoprolol), and CYP3A (midazolam) and was designed to overcome potential liabilities of other reported cocktails. Omeprazole 173-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 20002088-4 2009 * This cocktail can be used to test the effects of a new chemical entity on multiple CYP isoforms in a single clinical study: CYP1A2 (caffeine), CYP2C9 (warfarin), CYP2C19 (omeprazole), CYP2D6 (metoprolol), and CYP3A (midazolam) and was designed to overcome potential liabilities of other reported cocktails. Metoprolol 194-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 20002088-4 2009 * This cocktail can be used to test the effects of a new chemical entity on multiple CYP isoforms in a single clinical study: CYP1A2 (caffeine), CYP2C9 (warfarin), CYP2C19 (omeprazole), CYP2D6 (metoprolol), and CYP3A (midazolam) and was designed to overcome potential liabilities of other reported cocktails. Midazolam 218-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 20081260-4 2009 A comparative in vitro study provides clear evidence that ticlopidine and DDC, applied at concentrations that inhibit the above-mentioned CYP isoforms, potently (as compared to furafylline) inhibit human CYP1A2 and caffeine metabolism, in particular 1-N- and 3-N-demethylation. Ticlopidine 58-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 19616568-10 2009 This was further confirmed by significant inhibition of primaquine hemotoxicity by the selective CYP inhibitors, namely thiotepa (CYP2B6), fluoxetine (CYP2D6) and troleandomycin (CYP3A4). Fluoxetine 139-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 19616568-10 2009 This was further confirmed by significant inhibition of primaquine hemotoxicity by the selective CYP inhibitors, namely thiotepa (CYP2B6), fluoxetine (CYP2D6) and troleandomycin (CYP3A4). Troleandomycin 163-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 19616568-13 2009 The results provide significant insights regarding CYP isoforms contributing to hemotoxicity and may be useful in controlling toxicity of primaquine to increase its therapeutic utility. Primaquine 138-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 19523520-5 2009 Using the plant cytochrome P450 CYP79A1 and CYP71E1 as model systems, we demonstrate that a full-length CYP79A1 strepII tagged at the N-terminal expresses well and is able to translocate over the lipid bilayer to produce a functionally active protein that is amenable to affinity purification. Lipid Bilayers 196-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 19616568-0 2009 Cytochrome P(450)-dependent toxic effects of primaquine on human erythrocytes. Primaquine 45-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-17 19616568-9 2009 Multiple CYP isoforms (CYP2E1, CYP2B6, CYP1A2, CYP2D6 and CYP3A4) variably contributed to the hemotoxicity of primaquine. Primaquine 110-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 19616568-10 2009 This was further confirmed by significant inhibition of primaquine hemotoxicity by the selective CYP inhibitors, namely thiotepa (CYP2B6), fluoxetine (CYP2D6) and troleandomycin (CYP3A4). Primaquine 56-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 19616568-10 2009 This was further confirmed by significant inhibition of primaquine hemotoxicity by the selective CYP inhibitors, namely thiotepa (CYP2B6), fluoxetine (CYP2D6) and troleandomycin (CYP3A4). Thiotepa 120-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 20081260-4 2009 A comparative in vitro study provides clear evidence that ticlopidine and DDC, applied at concentrations that inhibit the above-mentioned CYP isoforms, potently (as compared to furafylline) inhibit human CYP1A2 and caffeine metabolism, in particular 1-N- and 3-N-demethylation. Ditiocarb 74-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 20081260-4 2009 A comparative in vitro study provides clear evidence that ticlopidine and DDC, applied at concentrations that inhibit the above-mentioned CYP isoforms, potently (as compared to furafylline) inhibit human CYP1A2 and caffeine metabolism, in particular 1-N- and 3-N-demethylation. Caffeine 215-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 20081260-4 2009 A comparative in vitro study provides clear evidence that ticlopidine and DDC, applied at concentrations that inhibit the above-mentioned CYP isoforms, potently (as compared to furafylline) inhibit human CYP1A2 and caffeine metabolism, in particular 1-N- and 3-N-demethylation. Nitrogen 252-253 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 19574470-2 2009 CONTENT: Tamoxifen requires enzymatic activation by cytochrome P450 (CYP) enzymes for the formation of active metabolites 4-hydroxytamoxifen and endoxifen. Tamoxifen 9-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 19534515-0 2009 Quantum mechanics/molecular mechanics studies on the sulfoxidation of dimethyl sulfide by compound I and compound 0 of cytochrome P450: which is the better oxidant? dimethyl sulfide 70-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-134 19788974-7 2009 RESULTS: Data demonstrate the presence, in some tested tissues, of cytochrome P-450 epoxygenase and soluble epoxide hydrolase, which respectively produce and degrade epoxyeicosatrienoic acid regioisomers. epoxyeicosatrienoic acid 166-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 19422358-4 2009 Probe drugs used to measure CYP activities were caffeine (CYP1A2), losartan (CYP2C9), omeprazole (CYP2C19), dextromethorphan (CYP2D6), chlorzoxazone (CYP2E1) and midazolam (CYP3A4). Caffeine 48-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 19198839-0 2009 Inhibition of drug metabolizing cytochrome P450s by the aromatase inhibitor drug letrozole and its major oxidative metabolite 4,4"-methanol-bisbenzonitrile in vitro. Letrozole 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 19198839-0 2009 Inhibition of drug metabolizing cytochrome P450s by the aromatase inhibitor drug letrozole and its major oxidative metabolite 4,4"-methanol-bisbenzonitrile in vitro. 4,4'-methanol-bisbenzonitrile 126-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 19198839-1 2009 PURPOSE: To determine the inhibitory potency of letrozole and its main human metabolite, 4,4"-methanol-bisbenzonitrile, on the activities of eight cytochrome P450 (CYP) enzymes. Letrozole 48-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-162 19198839-1 2009 PURPOSE: To determine the inhibitory potency of letrozole and its main human metabolite, 4,4"-methanol-bisbenzonitrile, on the activities of eight cytochrome P450 (CYP) enzymes. Letrozole 48-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-167 19198839-1 2009 PURPOSE: To determine the inhibitory potency of letrozole and its main human metabolite, 4,4"-methanol-bisbenzonitrile, on the activities of eight cytochrome P450 (CYP) enzymes. 4,4'-methanol-bisbenzonitrile 89-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-162 19198839-1 2009 PURPOSE: To determine the inhibitory potency of letrozole and its main human metabolite, 4,4"-methanol-bisbenzonitrile, on the activities of eight cytochrome P450 (CYP) enzymes. 4,4'-methanol-bisbenzonitrile 89-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-167 19198839-2 2009 METHODS: Letrozole and its metabolite were incubated with human liver microsomes (HLMs) (or expressed CYP isoforms) and NADPH in the absence (control) and presence of the test inhibitor. Letrozole 9-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 19574470-2 2009 CONTENT: Tamoxifen requires enzymatic activation by cytochrome P450 (CYP) enzymes for the formation of active metabolites 4-hydroxytamoxifen and endoxifen. Tamoxifen 9-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 19574470-2 2009 CONTENT: Tamoxifen requires enzymatic activation by cytochrome P450 (CYP) enzymes for the formation of active metabolites 4-hydroxytamoxifen and endoxifen. hydroxytamoxifen 122-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 19574470-2 2009 CONTENT: Tamoxifen requires enzymatic activation by cytochrome P450 (CYP) enzymes for the formation of active metabolites 4-hydroxytamoxifen and endoxifen. hydroxytamoxifen 122-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 19574470-2 2009 CONTENT: Tamoxifen requires enzymatic activation by cytochrome P450 (CYP) enzymes for the formation of active metabolites 4-hydroxytamoxifen and endoxifen. 4-hydroxy-N-desmethyltamoxifen 145-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 19574470-2 2009 CONTENT: Tamoxifen requires enzymatic activation by cytochrome P450 (CYP) enzymes for the formation of active metabolites 4-hydroxytamoxifen and endoxifen. 4-hydroxy-N-desmethyltamoxifen 145-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 19708688-0 2009 Kinetics and activation parameters for oxidations of styrene by Compounds I from the cytochrome P450(BM-3) (CYP102A1) heme domain and from CYP119. Styrene 53-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-105 19536146-3 2009 Here, we used normal human epidermal keratinocytes (NHEKs) to determine the effects of attainable levels of ozone exposure on the family of cytochrome P450 (CYP) isoforms, which plays a determinant role in the biotransformation of many environmental pollutants. Ozone 108-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-155 19536146-3 2009 Here, we used normal human epidermal keratinocytes (NHEKs) to determine the effects of attainable levels of ozone exposure on the family of cytochrome P450 (CYP) isoforms, which plays a determinant role in the biotransformation of many environmental pollutants. Ozone 108-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 19708688-0 2009 Kinetics and activation parameters for oxidations of styrene by Compounds I from the cytochrome P450(BM-3) (CYP102A1) heme domain and from CYP119. Heme 118-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-105 19708688-4 2009 We report here application of the photo-oxidation method for production of Compound I from the heme domain of CYP102A1 (cytochrome P450(BM-3)), and product and kinetic studies of reactions of styrene with this Compound I transient and also Compound I from CYP119. Heme 95-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-140 19708688-4 2009 We report here application of the photo-oxidation method for production of Compound I from the heme domain of CYP102A1 (cytochrome P450(BM-3)), and product and kinetic studies of reactions of styrene with this Compound I transient and also Compound I from CYP119. Styrene 192-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-140 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). Arachidonic Acid 81-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). Arachidonic Acid 81-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-24 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). epoxyeicosatrienenoic acids 120-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). epoxyeicosatrienenoic acids 120-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-24 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). eets 149-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). eets 149-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-24 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). hydroxyeicosatetrenoic acids 159-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). hydroxyeicosatetrenoic acids 159-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-24 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). hetes 189-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 19549792-1 2009 Cytochrome P-450 (CYP450) enzymes of the CYP2 and -4 family in humans metabolize arachidonic acid to generate bioactive epoxyeicosatrienenoic acids (EETs) and hydroxyeicosatetrenoic acids (HETEs). hetes 189-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-24 19593168-0 2009 ABCB1 and cytochrome P450 polymorphisms: clinical pharmacogenetics of clozapine. Clozapine 70-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 21217854-9 2009 Metabolic activation of Sudan I by both types of enzymes leads to formation of reactive species (the benzenediazonium ion by CYP and Sudan I radicals by peroxidase) that bind to DNA and RNA, generating covalent adducts in vitro and in vivo. 1-phenylazo-2-naphthol 24-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 21217854-9 2009 Metabolic activation of Sudan I by both types of enzymes leads to formation of reactive species (the benzenediazonium ion by CYP and Sudan I radicals by peroxidase) that bind to DNA and RNA, generating covalent adducts in vitro and in vivo. reactive species 79-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 21217854-9 2009 Metabolic activation of Sudan I by both types of enzymes leads to formation of reactive species (the benzenediazonium ion by CYP and Sudan I radicals by peroxidase) that bind to DNA and RNA, generating covalent adducts in vitro and in vivo. benzenediazonium 101-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 19480554-0 2009 Cytochrome P450-catalysed arene-epoxidation of the bioactive tea tree oil ingredient p-cymene: indication for the formation of a reactive allergenic intermediate? arene 26-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 19480554-0 2009 Cytochrome P450-catalysed arene-epoxidation of the bioactive tea tree oil ingredient p-cymene: indication for the formation of a reactive allergenic intermediate? 4-cymene 85-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 19480554-2 2009 The cytochrome P450-mediated metabolism of the tea tree oil ingredient p-cymene (p-isopropyltoluene) was studied by the application of in vitro enzymatic assays using different recombinant human cytochrome P450 enzymes. tree oil 51-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 19480554-2 2009 The cytochrome P450-mediated metabolism of the tea tree oil ingredient p-cymene (p-isopropyltoluene) was studied by the application of in vitro enzymatic assays using different recombinant human cytochrome P450 enzymes. tree oil 51-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-210 19480554-2 2009 The cytochrome P450-mediated metabolism of the tea tree oil ingredient p-cymene (p-isopropyltoluene) was studied by the application of in vitro enzymatic assays using different recombinant human cytochrome P450 enzymes. 4-cymene 71-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 19480554-2 2009 The cytochrome P450-mediated metabolism of the tea tree oil ingredient p-cymene (p-isopropyltoluene) was studied by the application of in vitro enzymatic assays using different recombinant human cytochrome P450 enzymes. 4-cymene 71-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-210 19480554-2 2009 The cytochrome P450-mediated metabolism of the tea tree oil ingredient p-cymene (p-isopropyltoluene) was studied by the application of in vitro enzymatic assays using different recombinant human cytochrome P450 enzymes. 4-cymene 81-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 19480554-2 2009 The cytochrome P450-mediated metabolism of the tea tree oil ingredient p-cymene (p-isopropyltoluene) was studied by the application of in vitro enzymatic assays using different recombinant human cytochrome P450 enzymes. 4-cymene 81-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-210 19480554-7 2009 The enzymatic products of p-cymene resulted from catalysed enzymatic arene-epoxidation and hydroxylation reactions by the studied cytochrome P450 enzymes. 4-cymene 26-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-145 19480554-7 2009 The enzymatic products of p-cymene resulted from catalysed enzymatic arene-epoxidation and hydroxylation reactions by the studied cytochrome P450 enzymes. arene 69-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-145 19480554-13 2009 The results also indicate that skin sensitization and irritation reactions not only can be explained by the frequently in literature reported auto-oxidation of tea tree resulting in bioactive oxidized products, but also now by the formation of epoxide intermediates resulting from catalysed arene-epoxidation reactions by selected human cytochrome P450 enzymes which are also located in different organs in humans. Epoxy Compounds 244-251 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 337-352 19480554-13 2009 The results also indicate that skin sensitization and irritation reactions not only can be explained by the frequently in literature reported auto-oxidation of tea tree resulting in bioactive oxidized products, but also now by the formation of epoxide intermediates resulting from catalysed arene-epoxidation reactions by selected human cytochrome P450 enzymes which are also located in different organs in humans. arene 291-296 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 337-352 19541489-2 2009 Highly oxidized lignans produced during the cytochrome P-450 metabolism in the cells show biological activities significantly different from those of their parent natural compounds. Lignans 16-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-60 19576320-0 2009 Relation of genetic polymorphisms in the cytochrome P450 gene with clopidogrel resistance after drug-eluting stent implantation in Koreans. Clopidogrel 67-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 19576320-1 2009 Clopidogrel is a prodrug that has to be converted to an active metabolite by hepatic cytochrome P450 (CYP) isoenzymes to inhibit platelet aggregation. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 19576320-1 2009 Clopidogrel is a prodrug that has to be converted to an active metabolite by hepatic cytochrome P450 (CYP) isoenzymes to inhibit platelet aggregation. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 19576320-3 2009 In this study, we sought to determine the relation of genetic polymorphisms of CYP genes to clopidogrel resistance in Koreans. Clopidogrel 92-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 19324859-0 2009 Expression of cytochrome P450 (CYP) enzymes in human nonpigmented ciliary epithelial cells: induction of CYP1B1 expression by TCDD. Polychlorinated Dibenzodioxins 126-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 19372226-4 2009 Cytochrome P450 (P450) 3A4 is the major isozyme involved in the oxidative biotransformation of motesanib, but the CYP2D6 and CYP1A isozymes also make minor contributions. motesanib diphosphate 95-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 19324859-0 2009 Expression of cytochrome P450 (CYP) enzymes in human nonpigmented ciliary epithelial cells: induction of CYP1B1 expression by TCDD. Polychlorinated Dibenzodioxins 126-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 19324859-1 2009 PURPOSE: Cytochrome P450 (CYP) enzymes metabolize endogenous compounds such as steroid hormones, fatty acids, and xenobiotics, including drugs and carcinogens. Steroids 79-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-24 19324859-1 2009 PURPOSE: Cytochrome P450 (CYP) enzymes metabolize endogenous compounds such as steroid hormones, fatty acids, and xenobiotics, including drugs and carcinogens. Steroids 79-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 19324859-1 2009 PURPOSE: Cytochrome P450 (CYP) enzymes metabolize endogenous compounds such as steroid hormones, fatty acids, and xenobiotics, including drugs and carcinogens. Fatty Acids 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-24 19324859-1 2009 PURPOSE: Cytochrome P450 (CYP) enzymes metabolize endogenous compounds such as steroid hormones, fatty acids, and xenobiotics, including drugs and carcinogens. Fatty Acids 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 19489632-6 2009 CYP3A4, CYP3A5, CYP3A7, and CYP19 (aromatase) inhibition to the log concentration of beta-acid content was significant with r(2) > 0.37, suggesting that these components can account for some of the variation in inhibition of CYP metabolism. 6-Hydroxy-2-oxo-1,2-dihydroquinoline-4-carboxylic acid 85-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 19496669-1 2009 OBJECTIVE: To investigate cytochrome P450 (CYP) enzymes involved in metabolism of racemic and S-ketamine in various species and to evaluate metabolic interactions of other analgesics with ketamine. Ketamine 94-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 19496669-1 2009 OBJECTIVE: To investigate cytochrome P450 (CYP) enzymes involved in metabolism of racemic and S-ketamine in various species and to evaluate metabolic interactions of other analgesics with ketamine. Ketamine 94-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 19496669-1 2009 OBJECTIVE: To investigate cytochrome P450 (CYP) enzymes involved in metabolism of racemic and S-ketamine in various species and to evaluate metabolic interactions of other analgesics with ketamine. Ketamine 96-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 19496669-1 2009 OBJECTIVE: To investigate cytochrome P450 (CYP) enzymes involved in metabolism of racemic and S-ketamine in various species and to evaluate metabolic interactions of other analgesics with ketamine. Ketamine 96-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 19496669-10 2009 CONCLUSIONS AND CLINICAL RELEVANCE: Enzymes of the CYP 3A4 family and orthologs of CYP 2C9 were involved in ketamine metabolism in horses, dogs, and humans. Ketamine 108-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 19480547-3 2009 Using this system, we confirmed that multiple CYP inactivation caused by mechanism-based inhibitors such as isoniazid and amiodarone could be detected simultaneously. Isoniazid 108-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 19473111-0 2009 Interactions between human cytochrome P450 enzymes and steroids: physiological and pharmacological implications. Steroids 55-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 19473111-2 2009 Human cytochrome P450 (CYP) enzymes present in probably every tissue are found responsible for biosynthesis and catabolism of steroids, which could result in either active or inactive metabolites. Steroids 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 19473111-2 2009 Human cytochrome P450 (CYP) enzymes present in probably every tissue are found responsible for biosynthesis and catabolism of steroids, which could result in either active or inactive metabolites. Steroids 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 19473111-3 2009 In addition, exposure to endogenous and exogenous steroids that causes modulation of CYP activities may substantially affect the pharmacokinetic behavior of a given drug. Steroids 50-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 19473111-4 2009 OBJECTIVE: This article summarizes our current understanding of the ability of steroids to act as substrates, inhibitors or heteroactivators for human CYP enzymes, with a specific focus on their functional consequences. Steroids 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 19473111-5 2009 METHODS: In the current review, we compare the mechanisms and regulation of CYP-mediated biotransformation of steroids, and in particular examine the diverse tissue distributions and biological roles of individual CYPs. Steroids 110-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 19480547-3 2009 Using this system, we confirmed that multiple CYP inactivation caused by mechanism-based inhibitors such as isoniazid and amiodarone could be detected simultaneously. Amiodarone 122-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 19330882-1 2009 Polycyclic aromatic hydrocarbons (PAHs) are activated by cytochrome P450 (CYP) isozymes, and a subset of the reactive metabolites generated is detoxified via conjugation with glutathione (GSH) by specific glutathione S-transferases (GSTs). Polycyclic Aromatic Hydrocarbons 0-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 19414633-0 2009 Cytochrome P450 genetic polymorphisms and the response to prasugrel: relationship to pharmacokinetic, pharmacodynamic, and clinical outcomes. Prasugrel Hydrochloride 58-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 19414633-1 2009 BACKGROUND: Both clopidogrel and prasugrel require biotransformation to active metabolites by cytochrome P450 (CYP) enzymes. Clopidogrel 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-109 19414633-1 2009 BACKGROUND: Both clopidogrel and prasugrel require biotransformation to active metabolites by cytochrome P450 (CYP) enzymes. Clopidogrel 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-114 19414633-1 2009 BACKGROUND: Both clopidogrel and prasugrel require biotransformation to active metabolites by cytochrome P450 (CYP) enzymes. Prasugrel Hydrochloride 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-109 19414633-1 2009 BACKGROUND: Both clopidogrel and prasugrel require biotransformation to active metabolites by cytochrome P450 (CYP) enzymes. Prasugrel Hydrochloride 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-114 19414633-3 2009 The effect of CYP polymorphisms on the clinical outcomes in patients treated with prasugrel remains unknown. Prasugrel Hydrochloride 82-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 19330882-1 2009 Polycyclic aromatic hydrocarbons (PAHs) are activated by cytochrome P450 (CYP) isozymes, and a subset of the reactive metabolites generated is detoxified via conjugation with glutathione (GSH) by specific glutathione S-transferases (GSTs). Polycyclic Aromatic Hydrocarbons 0-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 19330882-1 2009 Polycyclic aromatic hydrocarbons (PAHs) are activated by cytochrome P450 (CYP) isozymes, and a subset of the reactive metabolites generated is detoxified via conjugation with glutathione (GSH) by specific glutathione S-transferases (GSTs). Polycyclic Aromatic Hydrocarbons 34-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 19330882-1 2009 Polycyclic aromatic hydrocarbons (PAHs) are activated by cytochrome P450 (CYP) isozymes, and a subset of the reactive metabolites generated is detoxified via conjugation with glutathione (GSH) by specific glutathione S-transferases (GSTs). Polycyclic Aromatic Hydrocarbons 34-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 19330882-1 2009 Polycyclic aromatic hydrocarbons (PAHs) are activated by cytochrome P450 (CYP) isozymes, and a subset of the reactive metabolites generated is detoxified via conjugation with glutathione (GSH) by specific glutathione S-transferases (GSTs). Glutathione 175-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 19330882-1 2009 Polycyclic aromatic hydrocarbons (PAHs) are activated by cytochrome P450 (CYP) isozymes, and a subset of the reactive metabolites generated is detoxified via conjugation with glutathione (GSH) by specific glutathione S-transferases (GSTs). Glutathione 175-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 19330882-1 2009 Polycyclic aromatic hydrocarbons (PAHs) are activated by cytochrome P450 (CYP) isozymes, and a subset of the reactive metabolites generated is detoxified via conjugation with glutathione (GSH) by specific glutathione S-transferases (GSTs). Glutathione 188-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 19330882-1 2009 Polycyclic aromatic hydrocarbons (PAHs) are activated by cytochrome P450 (CYP) isozymes, and a subset of the reactive metabolites generated is detoxified via conjugation with glutathione (GSH) by specific glutathione S-transferases (GSTs). Glutathione 188-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 19426684-4 2009 These effects were associated with formation of two covalent ellipticine-derived DNA adducts, identical to those formed by the cytochrome P450- and peroxidase-mediated ellipticine metabolites, 13-hydroxy- and 12-hydroxyellipticine. ellipticine 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-158 19420747-4 2009 Here, we examined the induction of CYP gene expression by an inducer by examining the effect of phenobarbital treatment on CYP gene expression in the co-culture system. Phenobarbital 96-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 19420747-4 2009 Here, we examined the induction of CYP gene expression by an inducer by examining the effect of phenobarbital treatment on CYP gene expression in the co-culture system. Phenobarbital 96-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 19426684-4 2009 These effects were associated with formation of two covalent ellipticine-derived DNA adducts, identical to those formed by the cytochrome P450- and peroxidase-mediated ellipticine metabolites, 13-hydroxy- and 12-hydroxyellipticine. ellipticine 168-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-158 19426684-4 2009 These effects were associated with formation of two covalent ellipticine-derived DNA adducts, identical to those formed by the cytochrome P450- and peroxidase-mediated ellipticine metabolites, 13-hydroxy- and 12-hydroxyellipticine. 13-hydroxy- and 12-hydroxyellipticine 193-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-158 19030860-0 2009 Lenalidomide: in vitro evaluation of the metabolism and assessment of cytochrome P450 inhibition and induction. Lenalidomide 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 19030860-1 2009 PURPOSE: To assess the potential for drug-drug interactions between lenalidomide and substrates and inhibitors of cytochrome P450 (CYP) isozymes. Lenalidomide 68-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 19030860-3 2009 The inhibitory and inductive effects of lenalidomide on the CYP activities were evaluated in human liver microsomes and cultured human hepatocytes, respectively. Lenalidomide 40-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 19470220-3 2009 For studies on the quantitative involvement of human cytochrome P450 (CYP) isoenzymes in the initial hydroxylation, a reference standard of HO-MPHP was needed. ho-mphp 140-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 19519346-1 2009 Drug interactions occur frequently with triazole antifungal agents because of their properties as inhibitors of 1 or more phase 1 (cytochrome P450) biotransformation enzymes and, possibly, as inhibitors or substrates of a phase 2 biotransformation enzyme or transporter protein. Triazoles 40-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 19470220-3 2009 For studies on the quantitative involvement of human cytochrome P450 (CYP) isoenzymes in the initial hydroxylation, a reference standard of HO-MPHP was needed. ho-mphp 140-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 19470220-5 2009 MPHP.HNO(3) (250 micromol) was incubated with 1 L culture of the fission yeast (Schizosaccharomyces pombe) strain CAD64 heterologously co-expressing human CYP reductase and CYP2D6. Nitric Acid 5-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-158 19144770-1 2009 Phencyclidine (PCP) is a mechanism-based inactivator of cytochrome P450 (P450) 2B6. Phencyclidine 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-82 19245299-0 2009 In vitro and in vivo assessment of the effect of dalcetrapib on a panel of CYP substrates. dalcetrapib 49-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 19245299-1 2009 OBJECTIVE: The primary objective of this study was to investigate the drug-drug interaction potential of dalcetrapib on drugs metabolized via major cytochrome P450 (CYP) isoforms using both in vitro and clinical approaches. dalcetrapib 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-163 19245299-1 2009 OBJECTIVE: The primary objective of this study was to investigate the drug-drug interaction potential of dalcetrapib on drugs metabolized via major cytochrome P450 (CYP) isoforms using both in vitro and clinical approaches. dalcetrapib 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-168 19245299-6 2009 RESULTS: In vitro, dalcetrapib was inhibitory to all CYP enzymes tested. dalcetrapib 19-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 19245299-11 2009 CONCLUSIONS: Although in vitro studies indicated that dalcetrapib inhibits CYP activity, two clinical studies showed no clinically relevant effect on any of the major CYP isoforms at a 900 mg dose, which is higher than the 600 mg dose being explored in phase III studies. dalcetrapib 54-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 19601875-0 2009 Inhibitory properties of trapping agents: glutathione, potassium cyanide, and methoxylamine, against major human cytochrome p450 isoforms. Glutathione 42-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 19601875-0 2009 Inhibitory properties of trapping agents: glutathione, potassium cyanide, and methoxylamine, against major human cytochrome p450 isoforms. methoxyamine 78-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 19144770-1 2009 Phencyclidine (PCP) is a mechanism-based inactivator of cytochrome P450 (P450) 2B6. Phencyclidine 15-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-82 19241405-2 2009 Cytochrome P450 enzymes can also be used in artificial systems in which organic peroxides act as cosubstrates. Peroxides 80-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 19350451-0 2009 Characterization of human cytochrome P450 isoforms involved in the metabolism of 7-epi-paclitaxel. 7-epipaclitaxel 81-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 19350451-2 2009 This study was designed to characterize the cytochrome P450 (CYP) enzymes involved in 7-epi-paclitaxel metabolism, and to examine possible metabolic interactions that this C-7 epimer may have with paclitaxel. 7-epipaclitaxel 86-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 19350451-2 2009 This study was designed to characterize the cytochrome P450 (CYP) enzymes involved in 7-epi-paclitaxel metabolism, and to examine possible metabolic interactions that this C-7 epimer may have with paclitaxel. 7-epipaclitaxel 86-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 19350451-2 2009 This study was designed to characterize the cytochrome P450 (CYP) enzymes involved in 7-epi-paclitaxel metabolism, and to examine possible metabolic interactions that this C-7 epimer may have with paclitaxel. Paclitaxel 92-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 19350451-2 2009 This study was designed to characterize the cytochrome P450 (CYP) enzymes involved in 7-epi-paclitaxel metabolism, and to examine possible metabolic interactions that this C-7 epimer may have with paclitaxel. Paclitaxel 92-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 19196220-2 2009 Posaconazole, an extended-spectrum triazole, is an inhibitor of the cytochrome P450 (CYP) isoenzyme CYP3A4, and sirolimus, an immunosuppressant, is a substrate of the enzyme. posaconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 19414330-0 2009 Flexible structure of cytochrome P450: promiscuity of ligand binding in the CYP3A4 heme pocket. Heme 83-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 19196220-2 2009 Posaconazole, an extended-spectrum triazole, is an inhibitor of the cytochrome P450 (CYP) isoenzyme CYP3A4, and sirolimus, an immunosuppressant, is a substrate of the enzyme. posaconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 19196220-2 2009 Posaconazole, an extended-spectrum triazole, is an inhibitor of the cytochrome P450 (CYP) isoenzyme CYP3A4, and sirolimus, an immunosuppressant, is a substrate of the enzyme. Triazoles 35-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 19196220-2 2009 Posaconazole, an extended-spectrum triazole, is an inhibitor of the cytochrome P450 (CYP) isoenzyme CYP3A4, and sirolimus, an immunosuppressant, is a substrate of the enzyme. Triazoles 35-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 19196220-2 2009 Posaconazole, an extended-spectrum triazole, is an inhibitor of the cytochrome P450 (CYP) isoenzyme CYP3A4, and sirolimus, an immunosuppressant, is a substrate of the enzyme. Sirolimus 112-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 19337788-1 2009 The presence of cytochrome P450 (CYP) variant alleles may reduce the activation of the prodrug clopidogrel to its active state. Clopidogrel 95-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 19074527-1 2009 Cytochrome P450 (P450) 2B6 metabolizes a number of clinically relevant drugs and is one of the most highly polymorphic human P450 enzymes, with the Lys(262)-->Arg substitution being especially common in several genetic variants. Lysine 148-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 19074527-1 2009 Cytochrome P450 (P450) 2B6 metabolizes a number of clinically relevant drugs and is one of the most highly polymorphic human P450 enzymes, with the Lys(262)-->Arg substitution being especially common in several genetic variants. Arginine 162-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). Flavonoids 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). Flavones 134-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). Flavones 134-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). 6-hydroxyflavone 153-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). 7-hydroxyflavone 171-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). chrysin 189-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). baicalein 198-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). Apigenin 209-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). Luteolin 219-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). scutellarein 229-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). wogonin 247-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). Flavonols 260-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). fisetin 281-288 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). kaempferol 290-300 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19074529-1 2009 This article describes an in vitro investigation of the inhibition of cytochrome P450 (P450) 2C9 by a series of flavonoids made up of flavones (flavone, 6-hydroxyflavone, 7-hydroxyflavone, chrysin, baicalein, apigenin, luteolin, scutellarein, and wogonin) and flavonols (galangin, fisetin, kaempferol, morin, and quercetin). Quercetin 313-322 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-96 19337788-1 2009 The presence of cytochrome P450 (CYP) variant alleles may reduce the activation of the prodrug clopidogrel to its active state. Clopidogrel 95-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 19022226-0 2009 Investigations on the cytochrome P450 (CYP) isoenzymes involved in the metabolism of the designer drugs N-(1-phenyl cyclohexyl)-2-ethoxyethanamine and N-(1-phenylcyclohexyl)-2-methoxyethanamine. N-(1-phenylcyclohexyl)-2-ethoxyethanamine 104-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 19029318-0 2009 Comprehensive in vitro analysis of voriconazole inhibition of eight cytochrome P450 (CYP) enzymes: major effect on CYPs 2B6, 2C9, 2C19, and 3A. Voriconazole 35-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 19029318-0 2009 Comprehensive in vitro analysis of voriconazole inhibition of eight cytochrome P450 (CYP) enzymes: major effect on CYPs 2B6, 2C9, 2C19, and 3A. Voriconazole 35-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 19029318-2 2009 To identify the mechanisms of these interactions, we tested the inhibitory potency of voriconazole with eight human cytochrome P450 (CYP) enzymes. Voriconazole 86-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 19029318-2 2009 To identify the mechanisms of these interactions, we tested the inhibitory potency of voriconazole with eight human cytochrome P450 (CYP) enzymes. Voriconazole 86-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 19022226-0 2009 Investigations on the cytochrome P450 (CYP) isoenzymes involved in the metabolism of the designer drugs N-(1-phenyl cyclohexyl)-2-ethoxyethanamine and N-(1-phenylcyclohexyl)-2-methoxyethanamine. N-(1-phenylcyclohexyl)-2-ethoxyethanamine 104-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 19022226-0 2009 Investigations on the cytochrome P450 (CYP) isoenzymes involved in the metabolism of the designer drugs N-(1-phenyl cyclohexyl)-2-ethoxyethanamine and N-(1-phenylcyclohexyl)-2-methoxyethanamine. N-(1-phenylcyclohexyl)-2-methoxyethanamine 151-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 19022226-0 2009 Investigations on the cytochrome P450 (CYP) isoenzymes involved in the metabolism of the designer drugs N-(1-phenyl cyclohexyl)-2-ethoxyethanamine and N-(1-phenylcyclohexyl)-2-methoxyethanamine. N-(1-phenylcyclohexyl)-2-methoxyethanamine 151-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 18978038-1 2009 Human cytochrome P-450 epoxygenase enzymes metabolize eicosapentaenoic acid (EPA), an omega-3-polyunsaturated fatty acid (PUFA), and leads to the production of 17(18)-epoxyeicosatetraenoic acid, or 17(18)-EpETE. Eicosapentaenoic Acid 54-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 19182369-1 2009 Our earlier investigations demonstrated the remarkable activation of cytochrome P-450 reductase and nitric oxide synthase by 7,8-diacetoxy-4-methylcoumarin, a model polyphenolic acetate by way of acetylation, catalyzed by the Calreticulin. 7,8-diacetoxy-4-methylcoumarin 125-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 19182369-1 2009 Our earlier investigations demonstrated the remarkable activation of cytochrome P-450 reductase and nitric oxide synthase by 7,8-diacetoxy-4-methylcoumarin, a model polyphenolic acetate by way of acetylation, catalyzed by the Calreticulin. polyphenolic acetate 165-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 19004845-4 2009 Published data and data obtained from the drug"s manufacturer implies that the dose escalation after 48 hours is to compensate for fomepizole-induced increased body clearance resulting from autoinduction of the cytochrome P450 (CYP) drug metabolizing enzyme CYP2E1. Fomepizole 131-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-226 19004845-4 2009 Published data and data obtained from the drug"s manufacturer implies that the dose escalation after 48 hours is to compensate for fomepizole-induced increased body clearance resulting from autoinduction of the cytochrome P450 (CYP) drug metabolizing enzyme CYP2E1. Fomepizole 131-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-231 19029204-0 2009 Induction of CYP4F3 by benzene metabolites in human white blood cells in vivo in human promyelocytic leukemic cell lines and ex vivo in human blood neutrophils. Benzene 23-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-19 19029204-9 2009 Induction of CYP4F3 by phenol was also observed in differentiated HL-60 cells, in the proerythroid cell line K562, and ex vivo in human neutrophils. Phenol 23-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-19 19029204-11 2009 The findings demonstrated, for the first time, that benzene and metabolites induce CYP4F3 in human blood cells both in vivo and in vitro. Benzene 52-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-89 19029204-12 2009 Induction of CYP4F3 may play a role in the development of benzene hematotoxicity and serve as a biomarker of benzene exposure. Benzene 58-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-19 19029204-12 2009 Induction of CYP4F3 may play a role in the development of benzene hematotoxicity and serve as a biomarker of benzene exposure. Benzene 109-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-19 19106084-1 2009 BACKGROUND: Clopidogrel requires transformation into an active metabolite by cytochrome P-450 (CYP) enzymes for its antiplatelet effect. Clopidogrel 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-93 19106084-1 2009 BACKGROUND: Clopidogrel requires transformation into an active metabolite by cytochrome P-450 (CYP) enzymes for its antiplatelet effect. Clopidogrel 12-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-98 19106084-3 2009 METHODS: We tested the association between functional genetic variants in CYP genes, plasma concentrations of active drug metabolite, and platelet inhibition in response to clopidogrel in 162 healthy subjects. Clopidogrel 173-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 18978038-1 2009 Human cytochrome P-450 epoxygenase enzymes metabolize eicosapentaenoic acid (EPA), an omega-3-polyunsaturated fatty acid (PUFA), and leads to the production of 17(18)-epoxyeicosatetraenoic acid, or 17(18)-EpETE. Eicosapentaenoic Acid 77-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 18978038-1 2009 Human cytochrome P-450 epoxygenase enzymes metabolize eicosapentaenoic acid (EPA), an omega-3-polyunsaturated fatty acid (PUFA), and leads to the production of 17(18)-epoxyeicosatetraenoic acid, or 17(18)-EpETE. omega-3-polyunsaturated fatty acid 86-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 18978038-1 2009 Human cytochrome P-450 epoxygenase enzymes metabolize eicosapentaenoic acid (EPA), an omega-3-polyunsaturated fatty acid (PUFA), and leads to the production of 17(18)-epoxyeicosatetraenoic acid, or 17(18)-EpETE. pufa 122-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 18978038-1 2009 Human cytochrome P-450 epoxygenase enzymes metabolize eicosapentaenoic acid (EPA), an omega-3-polyunsaturated fatty acid (PUFA), and leads to the production of 17(18)-epoxyeicosatetraenoic acid, or 17(18)-EpETE. 17(18)-epoxyeicosatetraenoic acid 160-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 18978038-1 2009 Human cytochrome P-450 epoxygenase enzymes metabolize eicosapentaenoic acid (EPA), an omega-3-polyunsaturated fatty acid (PUFA), and leads to the production of 17(18)-epoxyeicosatetraenoic acid, or 17(18)-EpETE. 17,18-epoxy-5,8,11,14-eicosatetraenoic acid 198-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 19067475-10 2009 We suggest that inhibition of cytochrome P450 (CYP) enzymes 1A2 and 3A4 by ciprofloxacin resulted in delayed clozapine metabolism and elevated clozapine plasma concentrations. Ciprofloxacin 75-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 19133059-1 2009 AIMS: To investigate the influence of different cytochrome P450 (CYP) activities and other potential covariates on the disposition of methadone in patients on methadone maintenance therapy (MMT). Methadone 134-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 19133059-1 2009 AIMS: To investigate the influence of different cytochrome P450 (CYP) activities and other potential covariates on the disposition of methadone in patients on methadone maintenance therapy (MMT). Methadone 134-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 19133059-1 2009 AIMS: To investigate the influence of different cytochrome P450 (CYP) activities and other potential covariates on the disposition of methadone in patients on methadone maintenance therapy (MMT). mmt 190-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 19016711-1 2009 BACKGROUND: An alternative approach to retinoid therapy is to inhibit the cytochrome P450 (CYP)-mediated catabolism of endogenous all-trans retinoic acid in the skin by applying retinoic acid metabolism blocking agents such as talarozole (R115866). Retinoids 39-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 19016711-1 2009 BACKGROUND: An alternative approach to retinoid therapy is to inhibit the cytochrome P450 (CYP)-mediated catabolism of endogenous all-trans retinoic acid in the skin by applying retinoic acid metabolism blocking agents such as talarozole (R115866). Retinoids 39-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 19016711-1 2009 BACKGROUND: An alternative approach to retinoid therapy is to inhibit the cytochrome P450 (CYP)-mediated catabolism of endogenous all-trans retinoic acid in the skin by applying retinoic acid metabolism blocking agents such as talarozole (R115866). Tretinoin 130-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 19016711-1 2009 BACKGROUND: An alternative approach to retinoid therapy is to inhibit the cytochrome P450 (CYP)-mediated catabolism of endogenous all-trans retinoic acid in the skin by applying retinoic acid metabolism blocking agents such as talarozole (R115866). Tretinoin 130-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 19016711-1 2009 BACKGROUND: An alternative approach to retinoid therapy is to inhibit the cytochrome P450 (CYP)-mediated catabolism of endogenous all-trans retinoic acid in the skin by applying retinoic acid metabolism blocking agents such as talarozole (R115866). Tretinoin 140-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 19016711-1 2009 BACKGROUND: An alternative approach to retinoid therapy is to inhibit the cytochrome P450 (CYP)-mediated catabolism of endogenous all-trans retinoic acid in the skin by applying retinoic acid metabolism blocking agents such as talarozole (R115866). Tretinoin 140-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 19016711-1 2009 BACKGROUND: An alternative approach to retinoid therapy is to inhibit the cytochrome P450 (CYP)-mediated catabolism of endogenous all-trans retinoic acid in the skin by applying retinoic acid metabolism blocking agents such as talarozole (R115866). R 115866 227-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 19016711-1 2009 BACKGROUND: An alternative approach to retinoid therapy is to inhibit the cytochrome P450 (CYP)-mediated catabolism of endogenous all-trans retinoic acid in the skin by applying retinoic acid metabolism blocking agents such as talarozole (R115866). R 115866 227-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 19067475-10 2009 We suggest that inhibition of cytochrome P450 (CYP) enzymes 1A2 and 3A4 by ciprofloxacin resulted in delayed clozapine metabolism and elevated clozapine plasma concentrations. Ciprofloxacin 75-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 19067475-10 2009 We suggest that inhibition of cytochrome P450 (CYP) enzymes 1A2 and 3A4 by ciprofloxacin resulted in delayed clozapine metabolism and elevated clozapine plasma concentrations. Clozapine 109-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 19067475-10 2009 We suggest that inhibition of cytochrome P450 (CYP) enzymes 1A2 and 3A4 by ciprofloxacin resulted in delayed clozapine metabolism and elevated clozapine plasma concentrations. Clozapine 109-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 19067475-10 2009 We suggest that inhibition of cytochrome P450 (CYP) enzymes 1A2 and 3A4 by ciprofloxacin resulted in delayed clozapine metabolism and elevated clozapine plasma concentrations. Clozapine 143-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 19067475-10 2009 We suggest that inhibition of cytochrome P450 (CYP) enzymes 1A2 and 3A4 by ciprofloxacin resulted in delayed clozapine metabolism and elevated clozapine plasma concentrations. Clozapine 143-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 19127490-3 2009 Unlike various other antidepressants including venlafaxine, desvenlafaxine is not metabolized by cytochrome p450 (CYP) enzyme pathways and is associated with minimal inhibition of CYP enzymes. Desvenlafaxine Succinate 60-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 18838506-1 2009 In vitro experiments were conducted to compare k(inact), K(I) and inactivation efficiency (k(inact)/K(I)) of cytochrome P450 (P450) 2C9 by tienilic acid and (+/-)-suprofen using (S)-flurbiprofen, diclofenac, and (S)-warfarin as reporter substrates. Ticrynafen 139-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-135 18838506-1 2009 In vitro experiments were conducted to compare k(inact), K(I) and inactivation efficiency (k(inact)/K(I)) of cytochrome P450 (P450) 2C9 by tienilic acid and (+/-)-suprofen using (S)-flurbiprofen, diclofenac, and (S)-warfarin as reporter substrates. Flurbiprofen 178-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-135 18838506-1 2009 In vitro experiments were conducted to compare k(inact), K(I) and inactivation efficiency (k(inact)/K(I)) of cytochrome P450 (P450) 2C9 by tienilic acid and (+/-)-suprofen using (S)-flurbiprofen, diclofenac, and (S)-warfarin as reporter substrates. Diclofenac 196-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-135 18838506-1 2009 In vitro experiments were conducted to compare k(inact), K(I) and inactivation efficiency (k(inact)/K(I)) of cytochrome P450 (P450) 2C9 by tienilic acid and (+/-)-suprofen using (S)-flurbiprofen, diclofenac, and (S)-warfarin as reporter substrates. Warfarin 212-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-135 19252340-0 2009 Effect of hydrocotarnine on cytochrome P450 and P-glycoprotein. hydrocotarnine 10-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 19252340-2 2009 In this study, the effects of hydrocotarnine on the cytochrome P450 (CYP) and P-glycoprotein (P-gp) were investigated. hydrocotarnine 30-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 19566113-12 2009 Cinacalcet is extensively metabolized by multiple hepatic cytochrome P450 (CYP) enzymes (primarily 3A4, 2D6 and 1A2) with <1% of the parent drug excreted in the urine. Cinacalcet 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 19252340-2 2009 In this study, the effects of hydrocotarnine on the cytochrome P450 (CYP) and P-glycoprotein (P-gp) were investigated. hydrocotarnine 30-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 19566113-12 2009 Cinacalcet is extensively metabolized by multiple hepatic cytochrome P450 (CYP) enzymes (primarily 3A4, 2D6 and 1A2) with <1% of the parent drug excreted in the urine. Cinacalcet 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 19385713-1 2009 BACKGROUND AND OBJECTIVE: Bortezomib, an antineoplastic for the treatment of relapsed multiple myeloma and mantle cell lymphoma, undergoes metabolism through oxidative deboronation by cytochrome P450 (CYP) enzymes, primarily CYP3A4 and CYP2C19. Bortezomib 26-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-199 19385713-1 2009 BACKGROUND AND OBJECTIVE: Bortezomib, an antineoplastic for the treatment of relapsed multiple myeloma and mantle cell lymphoma, undergoes metabolism through oxidative deboronation by cytochrome P450 (CYP) enzymes, primarily CYP3A4 and CYP2C19. Bortezomib 26-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-204 19385713-4 2009 The variability of bortezomib pharmacokinetics with CYP enzyme polymorphism was also investigated. Bortezomib 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 19252340-6 2009 Furthermore, mice were intraperitoneally injected with hydrocotarnine for 14 days and the mRNA levels of major CYP isozymes and P-gp in the liver and small intestine were determined by real-time RT-PCR. hydrocotarnine 55-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-114 20408500-3 2009 Induction of CYP1A2, CYP2B6, CYP2C9, CYP2C19 and CYP3A4 by CGS (0.01, 0.3 and 3 mM) was evaluated in cryopreserved human hepatocytes, by determining CYP mRNA expression using quantitative RT-PCR. cysteinylglycine 59-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 19209593-0 2009 Chiral polychlorinated biphenyls are biotransformed enantioselectively by mammalian cytochrome P-450 isozymes to form hydroxylated metabolites. Polychlorinated Biphenyls 7-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 20408500-1 2009 The induction and inhibition of human hepatic cytochrome P450 (CYP) isoforms by crystalline glucosamine sulfate (CGS) was investigated in vitro. Glucosamine 92-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 20408500-1 2009 The induction and inhibition of human hepatic cytochrome P450 (CYP) isoforms by crystalline glucosamine sulfate (CGS) was investigated in vitro. Glucosamine 92-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 20408500-1 2009 The induction and inhibition of human hepatic cytochrome P450 (CYP) isoforms by crystalline glucosamine sulfate (CGS) was investigated in vitro. cysteinylglycine 113-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 20408500-1 2009 The induction and inhibition of human hepatic cytochrome P450 (CYP) isoforms by crystalline glucosamine sulfate (CGS) was investigated in vitro. cysteinylglycine 113-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 18848727-0 2009 Quantum chemical studies for oxidation of morpholine by Cytochrome P450. morpholine 42-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 18815777-0 2009 Cytochrome P450 and gene activation--cholesterol elimination and regression of atherosclerosis: author reply to the commentary. Cholesterol 37-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 18984015-1 2009 Cytochrome P450 (P450) 2A6 is able to catalyze indole hydroxylation to form the blue dye indigo. indole 47-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 18984015-1 2009 Cytochrome P450 (P450) 2A6 is able to catalyze indole hydroxylation to form the blue dye indigo. Indigo Carmine 89-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 18848727-1 2009 Since morpholine oxidation has recently been shown to involve Cytochrome P450, the study on its mechanism at molecular level using quantum chemical calculations for the model of cytochrome active site is reported here. morpholine 6-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 18992797-1 2008 The environmental carcinogen 5-methylchrysene (5MC) can be activated to mutagenic metabolites by several isozymes of cytochrome P-450 (CYP). 5-methylchrysene 29-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 18779756-1 2009 OBJECTIVE: The genetic variations of the genes encoding cytochrome P-450 enzymes are considered to play an important role in the metabolism of estradiol. Estradiol 143-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 20027147-1 2009 OBJECTIVES: The aim of this study was to assess the effect of various flavonoids on the NADPH:cytochrome P450 oxidoreductase (CYPOR) activity in respect of the reduction of different electron acceptors as well as to study an impact of flavonoids on monooxygenation of a model substrate of cytochrome P450 (CYP). Flavonoids 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 20027147-1 2009 OBJECTIVES: The aim of this study was to assess the effect of various flavonoids on the NADPH:cytochrome P450 oxidoreductase (CYPOR) activity in respect of the reduction of different electron acceptors as well as to study an impact of flavonoids on monooxygenation of a model substrate of cytochrome P450 (CYP). Flavonoids 235-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-109 20027147-1 2009 OBJECTIVES: The aim of this study was to assess the effect of various flavonoids on the NADPH:cytochrome P450 oxidoreductase (CYPOR) activity in respect of the reduction of different electron acceptors as well as to study an impact of flavonoids on monooxygenation of a model substrate of cytochrome P450 (CYP). Flavonoids 235-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 20027147-5 2009 RESULTS: The stimulation of CYPOR activity via ANF was found to be associated with following electron acceptors: cytochrome c, potassium ferricyanide, cytochrome b5, but not with CYP. potassium ferricyanide 127-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 19629022-6 2009 CONCLUSIONS: Considering in vitro and available clinical data, desvenlafaxine and venlafaxine appear to have low potential for pharmacokinetic drug-drug interactions via inhibiting the metabolic clearance of concomitant drugs that are substrates of various CYP enzymes, in particular CYP2D6. Desvenlafaxine Succinate 63-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 257-260 19629022-6 2009 CONCLUSIONS: Considering in vitro and available clinical data, desvenlafaxine and venlafaxine appear to have low potential for pharmacokinetic drug-drug interactions via inhibiting the metabolic clearance of concomitant drugs that are substrates of various CYP enzymes, in particular CYP2D6. Venlafaxine Hydrochloride 66-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 257-260 19495700-3 2009 This chapter describes methods for the measurement of the expression and catalytic activity of three key cytochrome P450 (CYP) enzymes involved in the production of progesterone and estrogens, aromatase (CYP19), steroid 17-hydroxylase/17,20-lyase (CYP17), and cholesterol side-chain cleavage (CYP11A). Progesterone 165-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 19495700-3 2009 This chapter describes methods for the measurement of the expression and catalytic activity of three key cytochrome P450 (CYP) enzymes involved in the production of progesterone and estrogens, aromatase (CYP19), steroid 17-hydroxylase/17,20-lyase (CYP17), and cholesterol side-chain cleavage (CYP11A). Progesterone 165-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 19495700-3 2009 This chapter describes methods for the measurement of the expression and catalytic activity of three key cytochrome P450 (CYP) enzymes involved in the production of progesterone and estrogens, aromatase (CYP19), steroid 17-hydroxylase/17,20-lyase (CYP17), and cholesterol side-chain cleavage (CYP11A). Cholesterol 260-271 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 19495700-3 2009 This chapter describes methods for the measurement of the expression and catalytic activity of three key cytochrome P450 (CYP) enzymes involved in the production of progesterone and estrogens, aromatase (CYP19), steroid 17-hydroxylase/17,20-lyase (CYP17), and cholesterol side-chain cleavage (CYP11A). Cholesterol 260-271 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 20027147-0 2009 Modulation of cytochrome P450 enzyme system by selected flavonoids. Flavonoids 56-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 20027147-1 2009 OBJECTIVES: The aim of this study was to assess the effect of various flavonoids on the NADPH:cytochrome P450 oxidoreductase (CYPOR) activity in respect of the reduction of different electron acceptors as well as to study an impact of flavonoids on monooxygenation of a model substrate of cytochrome P450 (CYP). Flavonoids 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-109 19814645-8 2009 CYP testing revealed the patient to be homozygous for inactivating polymorphism CYP2C19*2, which is responsible for the metabolism of lansoprazole. Lansoprazole 134-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 19219743-0 2009 Characterization of human cytochrome P450 enzymes involved in the biotransformation of eperisone. eperisone 87-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 19219743-2 2009 The objective of this study was to characterize the metabolic pathway involved in the biotransformation of eperisone mediated by human cytochrome P450 (CYP) enzymes. eperisone 107-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 19219743-2 2009 The objective of this study was to characterize the metabolic pathway involved in the biotransformation of eperisone mediated by human cytochrome P450 (CYP) enzymes. eperisone 107-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-155 19219744-0 2009 Acetaminophen bioactivation by human cytochrome P450 enzymes and animal microsomes. Acetaminophen 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 19219744-3 2009 The key mechanism in the hepatotoxicity is cytochrome P450 (CYP)-catalysed formation of the reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI) that is capable of binding to cellular macromolecules and in that way an LC/MS liquid chromatography/mass spectrometry (LC/MS) method was developed to measure NAPQI formation by trapping it to reduced glutathione. N-acetyl-4-benzoquinoneimine 113-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 19219744-3 2009 The key mechanism in the hepatotoxicity is cytochrome P450 (CYP)-catalysed formation of the reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI) that is capable of binding to cellular macromolecules and in that way an LC/MS liquid chromatography/mass spectrometry (LC/MS) method was developed to measure NAPQI formation by trapping it to reduced glutathione. N-acetyl-4-benzoquinoneimine 113-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 19219744-3 2009 The key mechanism in the hepatotoxicity is cytochrome P450 (CYP)-catalysed formation of the reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI) that is capable of binding to cellular macromolecules and in that way an LC/MS liquid chromatography/mass spectrometry (LC/MS) method was developed to measure NAPQI formation by trapping it to reduced glutathione. N-acetyl-4-benzoquinoneimine 144-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 19219744-3 2009 The key mechanism in the hepatotoxicity is cytochrome P450 (CYP)-catalysed formation of the reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI) that is capable of binding to cellular macromolecules and in that way an LC/MS liquid chromatography/mass spectrometry (LC/MS) method was developed to measure NAPQI formation by trapping it to reduced glutathione. N-acetyl-4-benzoquinoneimine 144-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 19219744-3 2009 The key mechanism in the hepatotoxicity is cytochrome P450 (CYP)-catalysed formation of the reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI) that is capable of binding to cellular macromolecules and in that way an LC/MS liquid chromatography/mass spectrometry (LC/MS) method was developed to measure NAPQI formation by trapping it to reduced glutathione. N-acetyl-4-benzoquinoneimine 310-315 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 19219744-3 2009 The key mechanism in the hepatotoxicity is cytochrome P450 (CYP)-catalysed formation of the reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI) that is capable of binding to cellular macromolecules and in that way an LC/MS liquid chromatography/mass spectrometry (LC/MS) method was developed to measure NAPQI formation by trapping it to reduced glutathione. N-acetyl-4-benzoquinoneimine 310-315 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 19219744-3 2009 The key mechanism in the hepatotoxicity is cytochrome P450 (CYP)-catalysed formation of the reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI) that is capable of binding to cellular macromolecules and in that way an LC/MS liquid chromatography/mass spectrometry (LC/MS) method was developed to measure NAPQI formation by trapping it to reduced glutathione. Glutathione 352-363 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 19219744-3 2009 The key mechanism in the hepatotoxicity is cytochrome P450 (CYP)-catalysed formation of the reactive metabolite, N-acetyl-p-benzoquinone imine (NAPQI) that is capable of binding to cellular macromolecules and in that way an LC/MS liquid chromatography/mass spectrometry (LC/MS) method was developed to measure NAPQI formation by trapping it to reduced glutathione. Glutathione 352-363 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 19219744-4 2009 This method was used to determine the bioactivation of acetaminophen at two concentrations: 50 microM therapeutic and 1 mM toxic by using nine human recombinant CYP enzymes: CYP1A1, CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4; and with different microsomes from experimental animals. Acetaminophen 55-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 19219744-9 2009 This study suggests that CYP3A4 is the major CYP enzyme form catalysing acetaminophen oxidation to NAPQI in human liver. Acetaminophen 72-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 19219744-9 2009 This study suggests that CYP3A4 is the major CYP enzyme form catalysing acetaminophen oxidation to NAPQI in human liver. N-acetyl-4-benzoquinoneimine 99-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 19105833-1 2008 BACKGROUND: Cytochrome P450 (CYP) enzyme 2J2, an epoxygenase predominantly expressed in the heart, metabolizes arachidonic acid to biologically active eicosanoids. Arachidonic Acid 111-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 19105833-1 2008 BACKGROUND: Cytochrome P450 (CYP) enzyme 2J2, an epoxygenase predominantly expressed in the heart, metabolizes arachidonic acid to biologically active eicosanoids. Arachidonic Acid 111-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 19105833-1 2008 BACKGROUND: Cytochrome P450 (CYP) enzyme 2J2, an epoxygenase predominantly expressed in the heart, metabolizes arachidonic acid to biologically active eicosanoids. Eicosanoids 151-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 19105833-1 2008 BACKGROUND: Cytochrome P450 (CYP) enzyme 2J2, an epoxygenase predominantly expressed in the heart, metabolizes arachidonic acid to biologically active eicosanoids. Eicosanoids 151-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 19105833-4 2008 This association supports the vascular protective role of CYP-derived eicosanoids in cardiovascular disease. Eicosanoids 70-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 18938231-0 2008 2,5-Dimethoxyamphetamine-derived designer drugs: studies on the identification of cytochrome P450 (CYP) isoenzymes involved in formation of their main metabolites and on their capability to inhibit CYP2D6. 2,5-dimethoxyamphetamine 0-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 18938231-0 2008 2,5-Dimethoxyamphetamine-derived designer drugs: studies on the identification of cytochrome P450 (CYP) isoenzymes involved in formation of their main metabolites and on their capability to inhibit CYP2D6. 2,5-dimethoxyamphetamine 0-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-102 18992797-1 2008 The environmental carcinogen 5-methylchrysene (5MC) can be activated to mutagenic metabolites by several isozymes of cytochrome P-450 (CYP). 5-methylchrysene 29-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 18992797-1 2008 The environmental carcinogen 5-methylchrysene (5MC) can be activated to mutagenic metabolites by several isozymes of cytochrome P-450 (CYP). 5-methylchrysene 47-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 18992797-1 2008 The environmental carcinogen 5-methylchrysene (5MC) can be activated to mutagenic metabolites by several isozymes of cytochrome P-450 (CYP). 5-methylchrysene 47-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 18992797-4 2008 The parent compound 5MC was only 2-fold more cytotoxic in the CYP-expressing cell lines than in the V79MZ parental cell line, while 5MC-1,2-dihydrodiol was more than 30-fold more cytotoxic in CYP-transfected cells compared to V79MZ cells. 5-methylchrysene 20-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 18992797-4 2008 The parent compound 5MC was only 2-fold more cytotoxic in the CYP-expressing cell lines than in the V79MZ parental cell line, while 5MC-1,2-dihydrodiol was more than 30-fold more cytotoxic in CYP-transfected cells compared to V79MZ cells. 1,2-dihydro-1,2-dihydroxy-5-methylchrysene 132-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-195 18992797-5 2008 Cells co-expressing either hCYP1B1 or hCYP1A1 together with hGSTP1 were 2-fold less sensitive to 5MC or 5MC-1,2-diol cytotoxicity than their CYP-only parent lines. 5-methylchrysene 97-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 18992797-5 2008 Cells co-expressing either hCYP1B1 or hCYP1A1 together with hGSTP1 were 2-fold less sensitive to 5MC or 5MC-1,2-diol cytotoxicity than their CYP-only parent lines. 5mc-1,2-diol 104-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 18992797-10 2008 These studies show that the relative efficacy of protection by hGSTP1 against mutagenicity of 5MC or 5MC-1,2-diol is in part determined by the specific CYP pathway that catalyzes activation to the toxic or mutagenic metabolites. 5-methylchrysene 94-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-155 18992797-10 2008 These studies show that the relative efficacy of protection by hGSTP1 against mutagenicity of 5MC or 5MC-1,2-diol is in part determined by the specific CYP pathway that catalyzes activation to the toxic or mutagenic metabolites. 5mc-1,2-diol 101-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-155 19051152-3 2008 The high-profile withdrawal of drugs such as mibefradil from the market because of unfavorable drug-drug interaction profiles has focused efforts on screening for cytochrome P450 (CYP)-mediated drug interactions early in the discovery paradigm and on predicting the impact of inhibition on the in vivo situation. Mibefradil 45-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-178 19051152-3 2008 The high-profile withdrawal of drugs such as mibefradil from the market because of unfavorable drug-drug interaction profiles has focused efforts on screening for cytochrome P450 (CYP)-mediated drug interactions early in the discovery paradigm and on predicting the impact of inhibition on the in vivo situation. Mibefradil 45-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 18779363-3 2008 Here, we report the ability of 12(R)-hydroxy-5(Z),8(Z),10(E), 14(Z)-eicosatetraenoic acid [12(R)-HETE], an arachidonic acid metabolite produced by either a lipoxygenase or cytochrome P-450 pathway, to act as a potent indirect modulator of the AHR pathway. 12(r)-hydroxy-5 31-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 18955142-0 2008 Effects of prior oral contraceptive use and soy isoflavonoids on estrogen-metabolizing cytochrome P450 enzymes. isoflavonoids 48-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 18955142-2 2008 In this study we investigated whether past oral contraceptive (OC) administration or current dietary isoflavonoids (IF) affected expression and/or activity of steroid hormone-metabolizing cytochrome P450 (CYP) enzymes using complementary primate and cell culture models. isoflavonoids 101-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-208 18955142-2 2008 In this study we investigated whether past oral contraceptive (OC) administration or current dietary isoflavonoids (IF) affected expression and/or activity of steroid hormone-metabolizing cytochrome P450 (CYP) enzymes using complementary primate and cell culture models. Steroids 159-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-203 18955142-2 2008 In this study we investigated whether past oral contraceptive (OC) administration or current dietary isoflavonoids (IF) affected expression and/or activity of steroid hormone-metabolizing cytochrome P450 (CYP) enzymes using complementary primate and cell culture models. Steroids 159-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-208 18955142-6 2008 For in vitro studies, we measured effects of the isoflavonoids genistein, daidzein and equol on CYP activity using intact V79 cells stably transfected to express CYP1A1, CYP1B1, or CYP3A4. isoflavonoids 49-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 18955142-6 2008 For in vitro studies, we measured effects of the isoflavonoids genistein, daidzein and equol on CYP activity using intact V79 cells stably transfected to express CYP1A1, CYP1B1, or CYP3A4. Genistein 63-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 18955142-6 2008 For in vitro studies, we measured effects of the isoflavonoids genistein, daidzein and equol on CYP activity using intact V79 cells stably transfected to express CYP1A1, CYP1B1, or CYP3A4. daidzein 74-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 18779363-3 2008 Here, we report the ability of 12(R)-hydroxy-5(Z),8(Z),10(E), 14(Z)-eicosatetraenoic acid [12(R)-HETE], an arachidonic acid metabolite produced by either a lipoxygenase or cytochrome P-450 pathway, to act as a potent indirect modulator of the AHR pathway. Arachidonic Acids 66-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 18779363-3 2008 Here, we report the ability of 12(R)-hydroxy-5(Z),8(Z),10(E), 14(Z)-eicosatetraenoic acid [12(R)-HETE], an arachidonic acid metabolite produced by either a lipoxygenase or cytochrome P-450 pathway, to act as a potent indirect modulator of the AHR pathway. Arachidonic Acid 107-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 18595692-1 2008 A derivative of rhodamine 110 has been designed and assessed as a probe for cytochrome P450 activity. rhodamine 110 16-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 18595692-4 2008 In cellulo, the probe revealed the induction of cytochrome P450 activity by the carcinogen 2,3,7,8-tetrachlorodibenzo-p-dioxin, and its repression by the chemoprotectant resveratrol. Polychlorinated Dibenzodioxins 91-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 18595692-4 2008 In cellulo, the probe revealed the induction of cytochrome P450 activity by the carcinogen 2,3,7,8-tetrachlorodibenzo-p-dioxin, and its repression by the chemoprotectant resveratrol. Resveratrol 170-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 18640101-6 2008 Honokiol efficiently scavenged superoxide radicals in xanthine oxidase and cytochrome P-450 cell-free systems with the rate constant 3.2x10(5)M(-1)s(-1), which is similar to reactivity of ascorbic acid but 20-times higher than reactivity of vitamin E analog trolox. honokiol 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 18787075-1 2008 Cytochrome P-450 (CYP) epoxygenases metabolize arachidonic acid to epoxyeicosatrienoic acid (EET) regioisomers, which activate several signaling pathways to promote endothelial cell proliferation, migration, and angiogenesis. Arachidonic Acid 47-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 18787075-1 2008 Cytochrome P-450 (CYP) epoxygenases metabolize arachidonic acid to epoxyeicosatrienoic acid (EET) regioisomers, which activate several signaling pathways to promote endothelial cell proliferation, migration, and angiogenesis. Arachidonic Acid 47-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 18787075-1 2008 Cytochrome P-450 (CYP) epoxygenases metabolize arachidonic acid to epoxyeicosatrienoic acid (EET) regioisomers, which activate several signaling pathways to promote endothelial cell proliferation, migration, and angiogenesis. epoxyeicosatrienoic acid 67-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 18787075-1 2008 Cytochrome P-450 (CYP) epoxygenases metabolize arachidonic acid to epoxyeicosatrienoic acid (EET) regioisomers, which activate several signaling pathways to promote endothelial cell proliferation, migration, and angiogenesis. epoxyeicosatrienoic acid 67-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 18585438-1 2008 Methoxychlor undergoes metabolism by cytochrome P450 (CYP) enzymes forming a chiral mono-phenolic derivative (Mono-OH-M) as main metabolite. Methoxychlor 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 18585438-1 2008 Methoxychlor undergoes metabolism by cytochrome P450 (CYP) enzymes forming a chiral mono-phenolic derivative (Mono-OH-M) as main metabolite. Methoxychlor 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 18634893-3 2008 METHODS: The assay consisted of human liver microsomes and a cocktail of probe substrates metabolized by the five major CYP isoforms (tacrine for CYP1A2, diclofenac for CYP2C9, (S)-mephenytoin for CYP2C19, dextromethorphan for CYP2D6 and midazolam for CYP3A4). Tacrine 134-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 18634893-3 2008 METHODS: The assay consisted of human liver microsomes and a cocktail of probe substrates metabolized by the five major CYP isoforms (tacrine for CYP1A2, diclofenac for CYP2C9, (S)-mephenytoin for CYP2C19, dextromethorphan for CYP2D6 and midazolam for CYP3A4). Diclofenac 154-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 18634893-3 2008 METHODS: The assay consisted of human liver microsomes and a cocktail of probe substrates metabolized by the five major CYP isoforms (tacrine for CYP1A2, diclofenac for CYP2C9, (S)-mephenytoin for CYP2C19, dextromethorphan for CYP2D6 and midazolam for CYP3A4). Dextromethorphan 206-222 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 18634893-3 2008 METHODS: The assay consisted of human liver microsomes and a cocktail of probe substrates metabolized by the five major CYP isoforms (tacrine for CYP1A2, diclofenac for CYP2C9, (S)-mephenytoin for CYP2C19, dextromethorphan for CYP2D6 and midazolam for CYP3A4). Midazolam 238-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 18566475-1 2008 The regulation of the human liver-specific cytochrome P450 4F3B (CYP4F3B) isoform, a splice variant of the CYP4F3 gene with strong substrate specificity for long chain fatty acids, is yet an unsolved question. long chain fatty acids 157-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-71 18566475-5 2008 SiRNA-mediated-silencing of CYP4F3 suppresses both 20-HETE synthesis and PGA1 induced 20-HETE production. 20-Hete 51-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-34 18566475-5 2008 SiRNA-mediated-silencing of CYP4F3 suppresses both 20-HETE synthesis and PGA1 induced 20-HETE production. prostaglandin A1 73-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-34 18566475-5 2008 SiRNA-mediated-silencing of CYP4F3 suppresses both 20-HETE synthesis and PGA1 induced 20-HETE production. 20-Hete 86-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-34 18606824-0 2008 Identification of novel endogenous cytochrome p450 arachidonate metabolites with high affinity for cannabinoid receptors. Arachidonic Acid 51-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 18606824-2 2008 The released arachidonic acid can be metabolized by three enzymatic pathways: the cyclooxygenase pathway forming prostaglandins and thromboxanes, the lipoxygenase pathway generating leukotrienes and lipoxins, and the cytochrome P450 (cP450) pathway producing epoxyeicosatrienoic acids and hydroxyeicosatetraenoic acids. Arachidonic Acid 13-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 217-232 18606824-2 2008 The released arachidonic acid can be metabolized by three enzymatic pathways: the cyclooxygenase pathway forming prostaglandins and thromboxanes, the lipoxygenase pathway generating leukotrienes and lipoxins, and the cytochrome P450 (cP450) pathway producing epoxyeicosatrienoic acids and hydroxyeicosatetraenoic acids. Arachidonic Acid 13-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-239 18585438-1 2008 Methoxychlor undergoes metabolism by cytochrome P450 (CYP) enzymes forming a chiral mono-phenolic derivative (Mono-OH-M) as main metabolite. 1,1,1-trichloro-2-(4-hydroxyphenyl)-2-(4-methoxyphenyl)ethane 110-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 18585438-1 2008 Methoxychlor undergoes metabolism by cytochrome P450 (CYP) enzymes forming a chiral mono-phenolic derivative (Mono-OH-M) as main metabolite. 1,1,1-trichloro-2-(4-hydroxyphenyl)-2-(4-methoxyphenyl)ethane 110-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 18640101-6 2008 Honokiol efficiently scavenged superoxide radicals in xanthine oxidase and cytochrome P-450 cell-free systems with the rate constant 3.2x10(5)M(-1)s(-1), which is similar to reactivity of ascorbic acid but 20-times higher than reactivity of vitamin E analog trolox. Superoxides 31-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 18657024-3 2008 She was treated with rifabutin-the rifamycin with the least cytochrome P450 (CYP) enzyme induction-and therapeutic drug monitoring was performed so that target serum concentrations of all antimicrobial agents could be achieved. rifamycin SV 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 18633604-0 2008 Cytochrome P450 and gene activation--from pharmacology to cholesterol elimination and regression of atherosclerosis. Cholesterol 58-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 18687246-3 2008 Clopidogrel is metabolically activated by several hepatic cytochrome P450 (CYP) isoenzymes, including CYP1A2. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 18687246-3 2008 Clopidogrel is metabolically activated by several hepatic cytochrome P450 (CYP) isoenzymes, including CYP1A2. Clopidogrel 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 18803983-9 2008 Carbamazepine, phenytoin, phenobarbital, and primidone were found to have prominent cytochrome P450 (CYP) enzyme-induction effects, while valproic acid had an inhibitory effect. Carbamazepine 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 18803983-9 2008 Carbamazepine, phenytoin, phenobarbital, and primidone were found to have prominent cytochrome P450 (CYP) enzyme-induction effects, while valproic acid had an inhibitory effect. Carbamazepine 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 18803983-9 2008 Carbamazepine, phenytoin, phenobarbital, and primidone were found to have prominent cytochrome P450 (CYP) enzyme-induction effects, while valproic acid had an inhibitory effect. Phenytoin 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 18803983-9 2008 Carbamazepine, phenytoin, phenobarbital, and primidone were found to have prominent cytochrome P450 (CYP) enzyme-induction effects, while valproic acid had an inhibitory effect. Phenytoin 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 18803983-9 2008 Carbamazepine, phenytoin, phenobarbital, and primidone were found to have prominent cytochrome P450 (CYP) enzyme-induction effects, while valproic acid had an inhibitory effect. Phenobarbital 26-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 18803983-9 2008 Carbamazepine, phenytoin, phenobarbital, and primidone were found to have prominent cytochrome P450 (CYP) enzyme-induction effects, while valproic acid had an inhibitory effect. Phenobarbital 26-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 18803983-9 2008 Carbamazepine, phenytoin, phenobarbital, and primidone were found to have prominent cytochrome P450 (CYP) enzyme-induction effects, while valproic acid had an inhibitory effect. Primidone 45-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 18803983-9 2008 Carbamazepine, phenytoin, phenobarbital, and primidone were found to have prominent cytochrome P450 (CYP) enzyme-induction effects, while valproic acid had an inhibitory effect. Primidone 45-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 19356091-1 2008 Troglitazone (TGZ) induced hepatotoxicity has been linked to cytochrome P450 (CYP)-catalyzed reactive metabolite formation. Troglitazone 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 19356091-1 2008 Troglitazone (TGZ) induced hepatotoxicity has been linked to cytochrome P450 (CYP)-catalyzed reactive metabolite formation. Troglitazone 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 19356091-1 2008 Troglitazone (TGZ) induced hepatotoxicity has been linked to cytochrome P450 (CYP)-catalyzed reactive metabolite formation. Troglitazone 14-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 19356091-1 2008 Troglitazone (TGZ) induced hepatotoxicity has been linked to cytochrome P450 (CYP)-catalyzed reactive metabolite formation. Troglitazone 14-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 18606824-7 2008 This is the first evidence of a cytochrome P450-dependent arachidonate metabolite that can activate G-protein-coupled cell membrane receptors and suggests a functional link between the cytochrome P450 enzyme system and the endocannabinoid system. Arachidonic Acid 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 18606824-7 2008 This is the first evidence of a cytochrome P450-dependent arachidonate metabolite that can activate G-protein-coupled cell membrane receptors and suggests a functional link between the cytochrome P450 enzyme system and the endocannabinoid system. Arachidonic Acid 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-200 23604745-1 2008 TheAlternaria toxins alternariol (AOH), alternariol-9-methyl ether (AME), altenuene (ALT) and isoaltenuene (iALT) undergo extensive oxidative metabolism, but the cytochrome P450 (CYP) isoforms responsible for the reported hydroxylation reactions are yet unknown. alternariol 21-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 23604745-1 2008 TheAlternaria toxins alternariol (AOH), alternariol-9-methyl ether (AME), altenuene (ALT) and isoaltenuene (iALT) undergo extensive oxidative metabolism, but the cytochrome P450 (CYP) isoforms responsible for the reported hydroxylation reactions are yet unknown. alternariol 21-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-182 23604745-1 2008 TheAlternaria toxins alternariol (AOH), alternariol-9-methyl ether (AME), altenuene (ALT) and isoaltenuene (iALT) undergo extensive oxidative metabolism, but the cytochrome P450 (CYP) isoforms responsible for the reported hydroxylation reactions are yet unknown. alternariol monomethyl ether 40-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 23604745-1 2008 TheAlternaria toxins alternariol (AOH), alternariol-9-methyl ether (AME), altenuene (ALT) and isoaltenuene (iALT) undergo extensive oxidative metabolism, but the cytochrome P450 (CYP) isoforms responsible for the reported hydroxylation reactions are yet unknown. alternariol monomethyl ether 40-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-182 23604745-1 2008 TheAlternaria toxins alternariol (AOH), alternariol-9-methyl ether (AME), altenuene (ALT) and isoaltenuene (iALT) undergo extensive oxidative metabolism, but the cytochrome P450 (CYP) isoforms responsible for the reported hydroxylation reactions are yet unknown. altenuene 74-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 23604745-1 2008 TheAlternaria toxins alternariol (AOH), alternariol-9-methyl ether (AME), altenuene (ALT) and isoaltenuene (iALT) undergo extensive oxidative metabolism, but the cytochrome P450 (CYP) isoforms responsible for the reported hydroxylation reactions are yet unknown. altenuene 74-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-182 23604745-1 2008 TheAlternaria toxins alternariol (AOH), alternariol-9-methyl ether (AME), altenuene (ALT) and isoaltenuene (iALT) undergo extensive oxidative metabolism, but the cytochrome P450 (CYP) isoforms responsible for the reported hydroxylation reactions are yet unknown. Isoaltenuene 94-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 23604745-1 2008 TheAlternaria toxins alternariol (AOH), alternariol-9-methyl ether (AME), altenuene (ALT) and isoaltenuene (iALT) undergo extensive oxidative metabolism, but the cytochrome P450 (CYP) isoforms responsible for the reported hydroxylation reactions are yet unknown. Isoaltenuene 94-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-182 23604745-2 2008 In the present study, the activities of twelve human CYP isoforms for the hydroxylation of AOH, AME, ALT and iALT at different positions have been determined. alternariol 91-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 19040121-1 2008 Cytochrome P450 (CYP) is a heme-containing enzyme superfamily metabolizing a wide variety of xenobiotics, including drugs and carcinogens. Heme 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 19040121-1 2008 Cytochrome P450 (CYP) is a heme-containing enzyme superfamily metabolizing a wide variety of xenobiotics, including drugs and carcinogens. Heme 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 18648675-0 2008 Ruthenium catalyzed biomimetic oxidation in organic synthesis inspired by cytochrome P-450. Ruthenium 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 18648675-1 2008 Simulation of the function of cytochrome P-450 with low valent ruthenium complex catalysts leads to the discovery of biomimetic, catalytic oxidation of various substrates selectively under mild conditions. Ruthenium 63-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 18249473-5 2008 Apart from insights in important molecular properties for CYP inhibition, the present results may also guide further design of curcumin analogues with less susceptibility to drug-drug interactions. Curcumin 127-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 18657024-3 2008 She was treated with rifabutin-the rifamycin with the least cytochrome P450 (CYP) enzyme induction-and therapeutic drug monitoring was performed so that target serum concentrations of all antimicrobial agents could be achieved. rifamycin SV 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 18346045-0 2008 Inhibitory effects on cytochrome p450 enzymes of pentamidine and its amidoxime pro-drug. Pentamidine 49-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 18346045-0 2008 Inhibitory effects on cytochrome p450 enzymes of pentamidine and its amidoxime pro-drug. amidoxime 69-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 18399696-0 2008 SNP detection for cytochrome P450 alleles by target-assembled tandem oligonucleotide systems based on exciplexes. Oligonucleotides 69-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 19138946-3 2008 Genotoxicity may be caused by cytochrome P450 (CYP)-mediated oxidation of catechol estrogens to quinones that react with DNA to form depurinating estrogen-DNA adducts. catechol 74-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 19138946-3 2008 Genotoxicity may be caused by cytochrome P450 (CYP)-mediated oxidation of catechol estrogens to quinones that react with DNA to form depurinating estrogen-DNA adducts. catechol 74-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 19138946-3 2008 Genotoxicity may be caused by cytochrome P450 (CYP)-mediated oxidation of catechol estrogens to quinones that react with DNA to form depurinating estrogen-DNA adducts. Quinones 96-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 19138946-3 2008 Genotoxicity may be caused by cytochrome P450 (CYP)-mediated oxidation of catechol estrogens to quinones that react with DNA to form depurinating estrogen-DNA adducts. Quinones 96-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 19138946-6 2008 Resveratrol has anticancer effects in diverse in vitro and in vivo systems and is an AhR antagonist that decreases CYP expression but induces NQO1 expression. Resveratrol 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 18433732-5 2008 The omega-hydroxylation and inactivation of pro-inflammatory eicosanoids by members of the CYP4F subfamily and the association of the CYP4F2 and CYP4F3 genes with inflammatory celiac disease indicate an important role in the resolution of inflammation. Eicosanoids 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-151 19468476-11 2008 Rifampicin has to be avoided in allograft recipients as it activates cytochrome-P450 enzymes and thereby decreases the therapeutic levels of cyclosporine and prednisolone. Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 18388875-1 2008 The cytochrome P450 (CYP) gene superfamily comprises a large group of hemoproteins with diverse functions in steroid, lipid, and xenobiotic metabolism. Steroids 109-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 18388875-1 2008 The cytochrome P450 (CYP) gene superfamily comprises a large group of hemoproteins with diverse functions in steroid, lipid, and xenobiotic metabolism. Steroids 109-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 18520135-12 2008 Bound sulfur also affected redox regulation by modifying active thiol residues in some liver cytosol enzymes and effectively inhibited cytochrome P-450-dependent lipid peroxidation induced by CCl(4) and t-BuOOH. Sulfur 6-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 18493655-4 2008 In support of this idea, we found that adding FKBP blocks binding of FK506 to the common cytochrome P(450) enzyme CYP3A4 in vitro. Tacrolimus 69-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-106 18496131-1 2008 PURPOSE: The anticancer agent, cyclophosphamide, is metabolized by cytochrome P450 (CYP), glutathione S-transferase (GST) and aldehyde dehydrogenase (ALDH) enzymes. Cyclophosphamide 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 18496131-1 2008 PURPOSE: The anticancer agent, cyclophosphamide, is metabolized by cytochrome P450 (CYP), glutathione S-transferase (GST) and aldehyde dehydrogenase (ALDH) enzymes. Cyclophosphamide 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 18520135-12 2008 Bound sulfur also affected redox regulation by modifying active thiol residues in some liver cytosol enzymes and effectively inhibited cytochrome P-450-dependent lipid peroxidation induced by CCl(4) and t-BuOOH. Cefaclor 192-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 18570159-0 2008 Effect of butylated hydroxytoluene, curcumin, propyl gallate and thiabendazole on cytochrome P450 forms in cultured human hepatocytes. Thiabendazole 65-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Cholesterol 125-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18570159-2 2008 The objective of this study was to investigate the effects of four food chemicals, namely butylated hydroxytoluene (BHT), curcumin (CC), propyl gallate (PG) and thiabendazole (TB), on cytochrome P450 (CYP) forms in cultured human hepatocytes. Butylated Hydroxytoluene 90-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-199 18570159-2 2008 The objective of this study was to investigate the effects of four food chemicals, namely butylated hydroxytoluene (BHT), curcumin (CC), propyl gallate (PG) and thiabendazole (TB), on cytochrome P450 (CYP) forms in cultured human hepatocytes. Thiabendazole 161-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-199 18570159-11 2008 In summary, the results demonstrate that TB is a mixed inducer of CYP forms in human hepatocytes inducing CYP1A, CYP2B and CYP3A forms, whereas BHT is an inducer of CYP2B and CYP3A forms. Thiabendazole 41-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 18482659-2 2008 BACKGROUND: The cytochrome P450 (CYP)-dependent conversion of clopidogrel to its active metabolite may contribute to the variability in antiplatelet effect of clopidogrel. Clopidogrel 62-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 18482659-2 2008 BACKGROUND: The cytochrome P450 (CYP)-dependent conversion of clopidogrel to its active metabolite may contribute to the variability in antiplatelet effect of clopidogrel. Clopidogrel 62-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 18482659-2 2008 BACKGROUND: The cytochrome P450 (CYP)-dependent conversion of clopidogrel to its active metabolite may contribute to the variability in antiplatelet effect of clopidogrel. Clopidogrel 159-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 18482659-2 2008 BACKGROUND: The cytochrome P450 (CYP)-dependent conversion of clopidogrel to its active metabolite may contribute to the variability in antiplatelet effect of clopidogrel. Clopidogrel 159-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Cholesterol 125-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 300-303 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Bile Acids and Salts 138-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Oxysterols 152-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Oxysterols 152-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 300-303 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Fatty Acids 190-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Fatty Acids 190-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 300-303 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Prostaglandins 203-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Prostaglandins I 219-232 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Leukotrienes 234-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Leukotrienes 234-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 300-303 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Steroids 248-264 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Steroids 248-264 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 300-303 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Ketones 266-272 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. Ketones 266-272 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 300-303 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. vitamines a 281-292 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18646550-5 2008 CYP isoenzymes participate in metabolic pathways important for proper physiological functioning of the human organism, i.e.: cholesterol, bile acid and oxysterol biosynthesis; metabolism of fatty acids, prostaglandins, prostacyclins, leukotrienes, steroid hormones, ketone bodies, vitamines A and D. CYP isoenzymes participate in the metabolism of over 80% of drugs and other xenobiotic substances which can be present in the human organism. vitamines a 281-292 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 300-303 18402469-0 2008 Comparisons of (+/-)-benzo[a]pyrene-trans-7,8-dihydrodiol activation by human cytochrome P450 and aldo-keto reductase enzymes: effect of redox state and expression levels. benzo(a)pyrene 7,8-dihydrodiol 15-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-117 18473751-2 2008 The formation of hydroxyderivatives of tacrine is well-established step in the metabolization of this drug in liver by microsomal cytochrome P450 enzymes family. Tacrine 39-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-145 18451235-2 2008 EXPERIMENTAL DESIGN: Human microsomes were used to determine the cytochrome P450 enzyme(s) involved in the metabolism of ixabepilone. ixabepilone 121-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 18421623-7 2008 The UGT 1 and 2 families were involved in the glucuronidation, and several CYPs participated in the metabolism of febuxostat, suggesting that there is little possibility that the blood concentration of febuxostat varies widely even if febuxostat is concomitantly administered with drugs that inhibit CYP or UGT enzyme. Febuxostat 114-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 18356043-0 2008 Identification of the human cytochrome P450 enzymes involved in the in vitro biotransformation of lynestrenol and norethindrone. Lynestrenol 98-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 18356043-0 2008 Identification of the human cytochrome P450 enzymes involved in the in vitro biotransformation of lynestrenol and norethindrone. Norethindrone 114-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 18356043-1 2008 This study examined the cytochrome P450 (CYP) enzyme selectivity of in vitro bioactivation of lynestrenol to norethindrone and the further metabolism of norethindrone. Lynestrenol 94-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 18356043-1 2008 This study examined the cytochrome P450 (CYP) enzyme selectivity of in vitro bioactivation of lynestrenol to norethindrone and the further metabolism of norethindrone. Lynestrenol 94-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 18356043-1 2008 This study examined the cytochrome P450 (CYP) enzyme selectivity of in vitro bioactivation of lynestrenol to norethindrone and the further metabolism of norethindrone. Norethindrone 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 18356043-1 2008 This study examined the cytochrome P450 (CYP) enzyme selectivity of in vitro bioactivation of lynestrenol to norethindrone and the further metabolism of norethindrone. Norethindrone 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 18356043-1 2008 This study examined the cytochrome P450 (CYP) enzyme selectivity of in vitro bioactivation of lynestrenol to norethindrone and the further metabolism of norethindrone. Norethindrone 153-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 18356043-1 2008 This study examined the cytochrome P450 (CYP) enzyme selectivity of in vitro bioactivation of lynestrenol to norethindrone and the further metabolism of norethindrone. Norethindrone 153-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 18004755-1 2008 Cytochrome P450 (P450) 2J2 catalyzes epoxidation of arachidonic acid to eicosatrienoic acids, which are related to a variety of diseases such as coronary artery disease, hypertension, and carcinogenesis. Arachidonic Acid 52-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 18004755-1 2008 Cytochrome P450 (P450) 2J2 catalyzes epoxidation of arachidonic acid to eicosatrienoic acids, which are related to a variety of diseases such as coronary artery disease, hypertension, and carcinogenesis. formic acid 72-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 18421623-0 2008 In vitro drug-drug interaction studies with febuxostat, a novel non-purine selective inhibitor of xanthine oxidase: plasma protein binding, identification of metabolic enzymes and cytochrome P450 inhibition. Febuxostat 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-195 18421623-8 2008 Examination of the inhibitory effect of febuxostat on CYP enzymes suggests that febuxostat minimally inhibits the activities of any CYP. Febuxostat 40-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 18421623-8 2008 Examination of the inhibitory effect of febuxostat on CYP enzymes suggests that febuxostat minimally inhibits the activities of any CYP. Febuxostat 80-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 18421623-8 2008 Examination of the inhibitory effect of febuxostat on CYP enzymes suggests that febuxostat minimally inhibits the activities of any CYP. Febuxostat 80-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 17868191-1 2008 The arachidonic acid metabolizing CYP enzymes with prominent roles in vascular regulation are epoxygenases of the two gene family which generate epoxyeicosatrienoic acids. Arachidonic Acid 4-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 17868191-1 2008 The arachidonic acid metabolizing CYP enzymes with prominent roles in vascular regulation are epoxygenases of the two gene family which generate epoxyeicosatrienoic acids. epoxyeicosatrienoic acids 145-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 18311908-1 2008 Type II cytochrome P450 (CYP) ligands cause inhibition by direct coordination to the heme iron atom. Heme 85-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 26620942-0 2008 Transition-State Docking of Flunitrazepam and Progesterone in Cytochrome P450. Flunitrazepam 28-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 26620942-0 2008 Transition-State Docking of Flunitrazepam and Progesterone in Cytochrome P450. Progesterone 46-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 18311908-1 2008 Type II cytochrome P450 (CYP) ligands cause inhibition by direct coordination to the heme iron atom. Heme 85-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 18311908-1 2008 Type II cytochrome P450 (CYP) ligands cause inhibition by direct coordination to the heme iron atom. Iron 90-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 18311908-1 2008 Type II cytochrome P450 (CYP) ligands cause inhibition by direct coordination to the heme iron atom. Iron 90-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 18311908-3 2008 The approach to design compounds with diminished CYP inhibition is different depending on whether the compound binds (type II ligand) or not (type I ligand) to the iron atom of the heme group. Iron 164-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 18311908-3 2008 The approach to design compounds with diminished CYP inhibition is different depending on whether the compound binds (type II ligand) or not (type I ligand) to the iron atom of the heme group. Heme 181-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 18311908-4 2008 In this study, the structural characteristics of nitrogen-containing compounds, which bind to the iron atom in two CYP isoforms (CYP2C9 and CYP3A4), were investigated. Nitrogen 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 18311908-4 2008 In this study, the structural characteristics of nitrogen-containing compounds, which bind to the iron atom in two CYP isoforms (CYP2C9 and CYP3A4), were investigated. Iron 98-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 17996418-4 2008 Under conditions where COX2 activity is inhibited, arachidonic acid accumulates or is converted to eicosanoids via lipoxygenases and cytochrome P450 (CYP) epoxygenases. Arachidonic Acid 51-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 17996418-4 2008 Under conditions where COX2 activity is inhibited, arachidonic acid accumulates or is converted to eicosanoids via lipoxygenases and cytochrome P450 (CYP) epoxygenases. Arachidonic Acid 51-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 17996418-4 2008 Under conditions where COX2 activity is inhibited, arachidonic acid accumulates or is converted to eicosanoids via lipoxygenases and cytochrome P450 (CYP) epoxygenases. Eicosanoids 99-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 17996418-4 2008 Under conditions where COX2 activity is inhibited, arachidonic acid accumulates or is converted to eicosanoids via lipoxygenases and cytochrome P450 (CYP) epoxygenases. Eicosanoids 99-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 18048487-0 2008 Physical incorporation of NADPH-cytochrome P450 reductase and cytochrome P450 into phospholipid vesicles using glycocholate and Bio-Beads. Phospholipids 83-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 18048487-0 2008 Physical incorporation of NADPH-cytochrome P450 reductase and cytochrome P450 into phospholipid vesicles using glycocholate and Bio-Beads. Glycocholic Acid 111-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 18187347-8 2008 Cytochrome P450 enzyme inducing AEDs are most commonly associated with a negative impact on bone, but studies also suggest an effect of valproate. Valproic Acid 136-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 18065749-6 2008 CYP4F11 was the lone CYP4F/A enzyme that effectively oxidized 3-hydroxypalmitate. 3-hydroxypalmitate 62-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-5 18065749-0 2008 Omega oxidation of 3-hydroxy fatty acids by the human CYP4F gene subfamily enzyme CYP4F11. 3-hydroxy fatty acids 19-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-59 18065749-4 2008 Polyspecific CYP4F antibodies markedly inhibited microsomal omega-hydroxylation of 3-hydroxystearate (68%) and 3-hydroxypalmitate (99%), whereas CYP4A11 and CYP2E1 antibodies had little effect. 3-hydroxystearate 83-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-18 18065749-4 2008 Polyspecific CYP4F antibodies markedly inhibited microsomal omega-hydroxylation of 3-hydroxystearate (68%) and 3-hydroxypalmitate (99%), whereas CYP4A11 and CYP2E1 antibodies had little effect. 3-hydroxypalmitate 111-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-18 18338302-3 2008 The pathophysiology of renal toxicity in acetaminophen poisoning has been attributed to cytochrome P-450 mixed function oxidase isoenzymes present in the kidney, although other mechanisms have been elucidated, including the role of prostaglandin synthetase and N-deacetylase enzymes. Acetaminophen 41-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 17967930-12 2008 Cytochrome P450 inducers enhanced the conversion of benzo[a]pyrene to these metabolites without altering mRNA levels of major phenol-conjugating SULT forms (SULT1A1, SULT1A3, and SULT1B1). Benzo(a)pyrene 52-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 18274957-3 2008 After 60-min incubation in human liver microsomes with beta-nicotinamide adenine dinucleotide phosphate (NADPH) or uridine diphosphate glucuronic acid (UDPGA), the residual KBH-A40 was 90.6% +/- 5.1% and 28.9% +/- 2.0% (t(1/2) = 26 min), respectively, suggesting that KBH-A40 is likely predominantly metabolized by glucuronidation, rather than by cytochrome P450 (CYP)-mediated oxidation. NADP 55-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 347-362 18274957-3 2008 After 60-min incubation in human liver microsomes with beta-nicotinamide adenine dinucleotide phosphate (NADPH) or uridine diphosphate glucuronic acid (UDPGA), the residual KBH-A40 was 90.6% +/- 5.1% and 28.9% +/- 2.0% (t(1/2) = 26 min), respectively, suggesting that KBH-A40 is likely predominantly metabolized by glucuronidation, rather than by cytochrome P450 (CYP)-mediated oxidation. NADP 55-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 364-367 17980859-4 2008 Oxidative metabolism of 5-methoxy-N,N-diisopropyltryptamine (Foxy) by human liver microsomes and recombinant cytochrome P450 enzymes. 5-methoxy-N,N-diisopropyltryptamine 24-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 18076219-0 2008 Interaction profile of armodafinil with medications metabolized by cytochrome P450 enzymes 1A2, 3A4 and 2C19 in healthy subjects. Modafinil 23-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 18063021-2 2008 Disposition of carboplatin is determined by renal clearance, while the taxanes are metabolized by cytochrome P450 (CYP450) enzymes. Taxoids 71-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 18063021-2 2008 Disposition of carboplatin is determined by renal clearance, while the taxanes are metabolized by cytochrome P450 (CYP450) enzymes. Taxoids 71-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-121 18063021-3 2008 Although the majority of taxane metabolism occurs in the liver, recent data have shown that some solid tumors express CYP450 enzymes in the tumors themselves. taxane 25-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-124 18076219-2 2008 The objective of this article is to summarize the results of three clinical drug-interaction studies assessing the potential for drug interactions of armodafinil with agents metabolized by cytochrome P450 (CYP) enzymes 1A2, 3A4 and 2C19. Modafinil 150-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-204 18076219-2 2008 The objective of this article is to summarize the results of three clinical drug-interaction studies assessing the potential for drug interactions of armodafinil with agents metabolized by cytochrome P450 (CYP) enzymes 1A2, 3A4 and 2C19. Modafinil 150-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-209 18855665-1 2008 Cytochrome P450 (CYP) enzymes are a superfamily of heme containing proteins that catalyze xenobiotic metabolism phase I reactions. Heme 51-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 19026034-4 2008 Systemic triazoles are inhibitors of cytochrome P450 (CYP) isozymes, such as CYP3A4, CYP2C9 and CYP2C19, to varying degrees. Triazoles 9-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 19026034-4 2008 Systemic triazoles are inhibitors of cytochrome P450 (CYP) isozymes, such as CYP3A4, CYP2C9 and CYP2C19, to varying degrees. Triazoles 9-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 18781947-5 2008 Lasting recent years it has become clear that human skin cells express various CYP enzymes, including CYP26AI which is responsible for the metabolism of retinoic acid in skin cells. Tretinoin 153-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 18479171-22 2008 Therefore, clinically significant drug interactions between abacavir and drugs metabolized by CYP enzymes are unlikely. abacavir 60-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 18855665-1 2008 Cytochrome P450 (CYP) enzymes are a superfamily of heme containing proteins that catalyze xenobiotic metabolism phase I reactions. Heme 51-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 19326769-3 2008 The present study was designed to examine chlorpyrifos and DEET mediated induction of CYP isoforms and also to characterize their potential cytotoxic effects on primary human hepatocytes. Chlorpyrifos 42-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 19326769-5 2008 Chlorpyrifos and DEET also mediated the expression of CYP isoforms, particularly CYP3A4, CYP2B6 and CYP1A1, as shown by CYP3A4-specific protein expression, testosterone metabolism and CYP1Al-specific activity assays. Chlorpyrifos 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 19326769-5 2008 Chlorpyrifos and DEET also mediated the expression of CYP isoforms, particularly CYP3A4, CYP2B6 and CYP1A1, as shown by CYP3A4-specific protein expression, testosterone metabolism and CYP1Al-specific activity assays. Testosterone 156-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 19326769-7 2008 Therefore, DEET and chlorpyrifos mediated induction of CYP mRNA and functional CYP isoforms together with their cytotoxic potential in human hepatocytes suggests that exposure to chlorpyrifos and/or DEET should be considered in human health impact analysis. Chlorpyrifos 20-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 19326769-7 2008 Therefore, DEET and chlorpyrifos mediated induction of CYP mRNA and functional CYP isoforms together with their cytotoxic potential in human hepatocytes suggests that exposure to chlorpyrifos and/or DEET should be considered in human health impact analysis. Chlorpyrifos 20-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 19326769-7 2008 Therefore, DEET and chlorpyrifos mediated induction of CYP mRNA and functional CYP isoforms together with their cytotoxic potential in human hepatocytes suggests that exposure to chlorpyrifos and/or DEET should be considered in human health impact analysis. Chlorpyrifos 179-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 19326769-7 2008 Therefore, DEET and chlorpyrifos mediated induction of CYP mRNA and functional CYP isoforms together with their cytotoxic potential in human hepatocytes suggests that exposure to chlorpyrifos and/or DEET should be considered in human health impact analysis. Chlorpyrifos 179-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 18098061-2 2008 CYP3A4 was identified as the major enzyme and CYP2C8 as a minor enzyme responsible for mirodenafil N-dealkylation based on correlation analysis, inhibition studies, and cDNA-expressed CYP enzyme activities. mirodenafil 87-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 18094219-1 2008 Prasugrel, a thienopyridine prodrug, is hydrolyzed in vivo by esterases to a thiolactone followed by a single cytochrome P450 (CYP)-dependent step to an active metabolite that is a potent inhibitor of adenosine diphosphate-induced platelet aggregation. Prasugrel Hydrochloride 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 18094219-1 2008 Prasugrel, a thienopyridine prodrug, is hydrolyzed in vivo by esterases to a thiolactone followed by a single cytochrome P450 (CYP)-dependent step to an active metabolite that is a potent inhibitor of adenosine diphosphate-induced platelet aggregation. Prasugrel Hydrochloride 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 18094219-1 2008 Prasugrel, a thienopyridine prodrug, is hydrolyzed in vivo by esterases to a thiolactone followed by a single cytochrome P450 (CYP)-dependent step to an active metabolite that is a potent inhibitor of adenosine diphosphate-induced platelet aggregation. thienopyridine 13-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 18094219-1 2008 Prasugrel, a thienopyridine prodrug, is hydrolyzed in vivo by esterases to a thiolactone followed by a single cytochrome P450 (CYP)-dependent step to an active metabolite that is a potent inhibitor of adenosine diphosphate-induced platelet aggregation. thienopyridine 13-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 18094219-1 2008 Prasugrel, a thienopyridine prodrug, is hydrolyzed in vivo by esterases to a thiolactone followed by a single cytochrome P450 (CYP)-dependent step to an active metabolite that is a potent inhibitor of adenosine diphosphate-induced platelet aggregation. Adenosine Diphosphate 201-222 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 18094219-1 2008 Prasugrel, a thienopyridine prodrug, is hydrolyzed in vivo by esterases to a thiolactone followed by a single cytochrome P450 (CYP)-dependent step to an active metabolite that is a potent inhibitor of adenosine diphosphate-induced platelet aggregation. Adenosine Diphosphate 201-222 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 18685268-5 2008 There are several microbial mechanisms responsible for biodegradation of dioxins, including oxidative degradation by dioxygenase-containing aerobic bacteria, bacterial and fungal cytochrome P-450, fungal lignolytic enzymes, reductive dechlorination by anaerobic bacteria, and direct ether ring cleavage by fungi containing etherase-like enzymes. Dioxins 73-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-195 18589587-4 2008 A population with slow acetylation has a higher risk of toxicity, as that potent inhibition of cytochrome P450 (CYP450) isoforms by isoniazid (CYP2C19 y CYP3A) are dependent of INH plasmatic concentration. Isoniazid 132-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 18589587-4 2008 A population with slow acetylation has a higher risk of toxicity, as that potent inhibition of cytochrome P450 (CYP450) isoforms by isoniazid (CYP2C19 y CYP3A) are dependent of INH plasmatic concentration. Isoniazid 132-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-118 18098061-0 2008 Identification of cytochrome P450 enzymes responsible for N -dealkylation of a new oral erectogenic, mirodenafil. Nitrogen 58-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 18098061-0 2008 Identification of cytochrome P450 enzymes responsible for N -dealkylation of a new oral erectogenic, mirodenafil. mirodenafil 101-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 18098061-1 2008 The purpose of this paper is to characterize the cytochrome P450 (CYP) enzymes involved in the metabolism of a new oral erectogenic, mirodenafil, to a major circulating active metabolite, N-dehydroxyethyl-mirodenafil, and to investigate the inhibitory potential of mirodenafil on seven CYP enzymes in human liver microsomes. mirodenafil 133-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 18098061-1 2008 The purpose of this paper is to characterize the cytochrome P450 (CYP) enzymes involved in the metabolism of a new oral erectogenic, mirodenafil, to a major circulating active metabolite, N-dehydroxyethyl-mirodenafil, and to investigate the inhibitory potential of mirodenafil on seven CYP enzymes in human liver microsomes. mirodenafil 133-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 18176065-1 2008 We propose an improved method to measure urinary D-glucaric acid (GA), which might be of value as an indirect index of the activity of cytochrome P450 (CYP). Glucaric Acid 49-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 18098061-1 2008 The purpose of this paper is to characterize the cytochrome P450 (CYP) enzymes involved in the metabolism of a new oral erectogenic, mirodenafil, to a major circulating active metabolite, N-dehydroxyethyl-mirodenafil, and to investigate the inhibitory potential of mirodenafil on seven CYP enzymes in human liver microsomes. mirodenafil 133-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 286-289 18098061-1 2008 The purpose of this paper is to characterize the cytochrome P450 (CYP) enzymes involved in the metabolism of a new oral erectogenic, mirodenafil, to a major circulating active metabolite, N-dehydroxyethyl-mirodenafil, and to investigate the inhibitory potential of mirodenafil on seven CYP enzymes in human liver microsomes. N-dehydroxyethylmirodenafil 188-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 18098061-1 2008 The purpose of this paper is to characterize the cytochrome P450 (CYP) enzymes involved in the metabolism of a new oral erectogenic, mirodenafil, to a major circulating active metabolite, N-dehydroxyethyl-mirodenafil, and to investigate the inhibitory potential of mirodenafil on seven CYP enzymes in human liver microsomes. N-dehydroxyethylmirodenafil 188-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 18098061-1 2008 The purpose of this paper is to characterize the cytochrome P450 (CYP) enzymes involved in the metabolism of a new oral erectogenic, mirodenafil, to a major circulating active metabolite, N-dehydroxyethyl-mirodenafil, and to investigate the inhibitory potential of mirodenafil on seven CYP enzymes in human liver microsomes. N-dehydroxyethylmirodenafil 188-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 286-289 18176065-1 2008 We propose an improved method to measure urinary D-glucaric acid (GA), which might be of value as an indirect index of the activity of cytochrome P450 (CYP). Glucaric Acid 49-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-155 18098061-1 2008 The purpose of this paper is to characterize the cytochrome P450 (CYP) enzymes involved in the metabolism of a new oral erectogenic, mirodenafil, to a major circulating active metabolite, N-dehydroxyethyl-mirodenafil, and to investigate the inhibitory potential of mirodenafil on seven CYP enzymes in human liver microsomes. mirodenafil 205-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 18176065-1 2008 We propose an improved method to measure urinary D-glucaric acid (GA), which might be of value as an indirect index of the activity of cytochrome P450 (CYP). Glucaric Acid 66-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 18098061-1 2008 The purpose of this paper is to characterize the cytochrome P450 (CYP) enzymes involved in the metabolism of a new oral erectogenic, mirodenafil, to a major circulating active metabolite, N-dehydroxyethyl-mirodenafil, and to investigate the inhibitory potential of mirodenafil on seven CYP enzymes in human liver microsomes. mirodenafil 205-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 18098061-1 2008 The purpose of this paper is to characterize the cytochrome P450 (CYP) enzymes involved in the metabolism of a new oral erectogenic, mirodenafil, to a major circulating active metabolite, N-dehydroxyethyl-mirodenafil, and to investigate the inhibitory potential of mirodenafil on seven CYP enzymes in human liver microsomes. mirodenafil 205-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 286-289 18176065-1 2008 We propose an improved method to measure urinary D-glucaric acid (GA), which might be of value as an indirect index of the activity of cytochrome P450 (CYP). Glucaric Acid 66-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-155 17872452-2 2007 Here we address the mechanisms by which cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) contribute to this effect in native and cultured endothelial cells. epoxyeicosatrienoic acids 70-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 17872452-2 2007 Here we address the mechanisms by which cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) contribute to this effect in native and cultured endothelial cells. epoxyeicosatrienoic acids 70-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 17872452-2 2007 Here we address the mechanisms by which cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) contribute to this effect in native and cultured endothelial cells. eets 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 17872452-2 2007 Here we address the mechanisms by which cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) contribute to this effect in native and cultured endothelial cells. eets 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 17872452-3 2007 METHODS AND RESULTS: In native CYP2C-expressing endothelial cells, bradykinin elicited a Ca(2+) influx that was potentiated by the soluble epoxide hydrolase inhibitor, 1-adamantyl-3-cyclohexylurea (ACU), and attenuated by CYP inhibition. CHEMBL242255 168-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 17900275-3 2007 OBJECTIVE: To determine the relationship between genetic variation in cytochrome P450 (CYP) isoenzymes and the pharmacokinetic/pharmacodynamic response to prasugrel and clopidogrel. Prasugrel Hydrochloride 155-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 18220565-4 2007 Their formation is the result of hydrolysis (INA, HZ), cytochrome P450 (CYP)-dependent oxidation (HZ, NH(3), oxidizing free radicals), and N-acetyltransferase (NAT) activity (AcINH, AcHZ, DiAcHZ). Free Radicals 119-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 18034666-1 2007 The nonsteroidal anti-inflammatory drug diclofenac is extensively metabolized by cytochrome P450 (CYP) enzymes, mainly by CYP2C9. Diclofenac 40-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 18034666-1 2007 The nonsteroidal anti-inflammatory drug diclofenac is extensively metabolized by cytochrome P450 (CYP) enzymes, mainly by CYP2C9. Diclofenac 40-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 18034666-2 2007 Our objective was to study the effect of voriconazole, a potent inhibitor of several CYP enzymes, on the pharmacokinetics of diclofenac. Voriconazole 41-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 18034666-2 2007 Our objective was to study the effect of voriconazole, a potent inhibitor of several CYP enzymes, on the pharmacokinetics of diclofenac. Diclofenac 125-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 18034666-10 2007 In conclusion, voriconazole increased exposure to diclofenac, probably mainly by inhibition of its cytochrome P450 (CYP)-mediated metabolism. Voriconazole 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-114 18034666-10 2007 In conclusion, voriconazole increased exposure to diclofenac, probably mainly by inhibition of its cytochrome P450 (CYP)-mediated metabolism. Voriconazole 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 18034666-10 2007 In conclusion, voriconazole increased exposure to diclofenac, probably mainly by inhibition of its cytochrome P450 (CYP)-mediated metabolism. Diclofenac 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-114 18034666-10 2007 In conclusion, voriconazole increased exposure to diclofenac, probably mainly by inhibition of its cytochrome P450 (CYP)-mediated metabolism. Diclofenac 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 18220565-4 2007 Their formation is the result of hydrolysis (INA, HZ), cytochrome P450 (CYP)-dependent oxidation (HZ, NH(3), oxidizing free radicals), and N-acetyltransferase (NAT) activity (AcINH, AcHZ, DiAcHZ). Ammonia 102-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 18220565-4 2007 Their formation is the result of hydrolysis (INA, HZ), cytochrome P450 (CYP)-dependent oxidation (HZ, NH(3), oxidizing free radicals), and N-acetyltransferase (NAT) activity (AcINH, AcHZ, DiAcHZ). Ammonia 102-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 17900275-3 2007 OBJECTIVE: To determine the relationship between genetic variation in cytochrome P450 (CYP) isoenzymes and the pharmacokinetic/pharmacodynamic response to prasugrel and clopidogrel. Prasugrel Hydrochloride 155-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 17900275-3 2007 OBJECTIVE: To determine the relationship between genetic variation in cytochrome P450 (CYP) isoenzymes and the pharmacokinetic/pharmacodynamic response to prasugrel and clopidogrel. Clopidogrel 169-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 17900275-3 2007 OBJECTIVE: To determine the relationship between genetic variation in cytochrome P450 (CYP) isoenzymes and the pharmacokinetic/pharmacodynamic response to prasugrel and clopidogrel. Clopidogrel 169-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 18001838-4 2007 We present a review of the major polymorphic CYP alleles and conclude that this variability is of greatest importance for treatment with several antidepressants, antipsychotics, antiulcer drugs, anti-HIV drugs, anticoagulants, antidiabetics and the anticancer drug tamoxifen. Tamoxifen 265-274 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 18066112-10 2007 Our data indicate that both the aqueous and methanol extracts of all 5 uva-ursi products showed high cytochrome P450 isozyme inhibition, with the exception of the methanol extracts against cytochromes P3A4 and P19, which had low to moderate activity. Methanol 44-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 18033636-0 2007 Metabolism of CJ-036878, N-(3-phenethoxybenzyl)-4-hydroxybenzamide, in liver microsomes and recombinant cytochrome P450 enzymes: metabolite identification by LC-UV/MS(n) and (1)H-NMR. N-(3-phenethoxybenzyl)-4-hydroxybenzamide 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 18033636-2 2007 The present study investigated the phase I metabolism of CJ-036878 (N-(3-phenethoxybenzyl)-4-hydroxybenzamide), a potent antagonist of the N-methyl-D-asparatate (NMDA) receptor, using liver microsomes and representative recombinant cytochrome P450 enzymes. N-(3-phenethoxybenzyl)-4-hydroxybenzamide 57-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-247 17709371-3 2007 In the present study, the resistance of methoxylated flavones toward oxidative metabolism was investigated with human liver microsomes and recombinant cytochrome P450 (P450) isoforms. Flavones 53-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-182 17850846-3 2007 Of the stress-related genes, the expressions of the aryl hydrocarbon receptor (AhR), cytochrome P450 (CYP) and p53 genes were most significantly induced by exposure to PCDDs/DFs. Polychlorinated Dibenzodioxins 168-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 17850846-3 2007 Of the stress-related genes, the expressions of the aryl hydrocarbon receptor (AhR), cytochrome P450 (CYP) and p53 genes were most significantly induced by exposure to PCDDs/DFs. Polychlorinated Dibenzodioxins 168-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 17850846-3 2007 Of the stress-related genes, the expressions of the aryl hydrocarbon receptor (AhR), cytochrome P450 (CYP) and p53 genes were most significantly induced by exposure to PCDDs/DFs. aspartyl-phenylalanine 174-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 17850846-3 2007 Of the stress-related genes, the expressions of the aryl hydrocarbon receptor (AhR), cytochrome P450 (CYP) and p53 genes were most significantly induced by exposure to PCDDs/DFs. aspartyl-phenylalanine 174-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 17850846-6 2007 Statistical tests revealed significant correlations between the PCDDs/DFs concentration and the AhR and CYP gene expression/cell viability/DNA damage. Polychlorinated Dibenzodioxins 64-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-107 17850846-6 2007 Statistical tests revealed significant correlations between the PCDDs/DFs concentration and the AhR and CYP gene expression/cell viability/DNA damage. aspartyl-phenylalanine 70-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-107 17709372-0 2007 Human enteric microsomal CYP4F enzymes O-demethylate the antiparasitic prodrug pafuramidine. o-demethylate 39-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-30 17709372-0 2007 Human enteric microsomal CYP4F enzymes O-demethylate the antiparasitic prodrug pafuramidine. pafuramidine 79-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-30 17709372-1 2007 CYP4F enzymes, including CYP4F2 and CYP4F3B, were recently shown to be the major enzymes catalyzing the initial oxidative O-demethylation of the antiparasitic prodrug pafuramidine (DB289) by human liver microsomes. pafuramidine 167-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-5 21783812-10 2007 This suggests that NMPA formation is partially dependent on cytochrome-P450 reductase. N-methyl-N-2-(methylsulfinyl)ethylpropionic acid amide 19-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 17709372-1 2007 CYP4F enzymes, including CYP4F2 and CYP4F3B, were recently shown to be the major enzymes catalyzing the initial oxidative O-demethylation of the antiparasitic prodrug pafuramidine (DB289) by human liver microsomes. pafuramidine 181-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-5 17709372-11 2007 We conclude that enteric CYP4F enzymes could play a role in the first-pass biotransformation of DB289 and other xenobiotics. pafuramidine 96-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-30 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). 6beta-hydroxy 95-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 17693640-3 2007 Approximately 500 pmol of norbenzphetamine and 58 pmol of cyclohexanol were formed per nmol of cyt P450. norbenzphetamine 26-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-103 17693640-3 2007 Approximately 500 pmol of norbenzphetamine and 58 pmol of cyclohexanol were formed per nmol of cyt P450. Cyclohexanols 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-103 17693640-5 2007 Four hundred picomoles of norbenzphetamine and 21 pmol of cyclohexanol were formed per nmol of cyt P450. norbenzphetamine 26-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-103 17693640-5 2007 Four hundred picomoles of norbenzphetamine and 21 pmol of cyclohexanol were formed per nmol of cyt P450. Cyclohexanols 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-103 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). 7beta-ohgz 138-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). 7beta-ohgz 138-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). 6beta-hydroxy 95-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). meoh-gz 169-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). 6beta-ohgz 110-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). meoh-gz 169-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). gliclizide 193-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). 6beta-ohgz 110-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). gliclizide 193-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). 7beta-hydroxy 123-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). gz 118-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). gz 118-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 17517049-1 2007 AIMS: To identify the human cytochrome P450 (CYP) enzymes responsible for the formation of the 6beta-hydroxy (6beta-OHGz), 7beta-hydroxy (7beta-OHGz) and hydroxymethyl (MeOH-Gz) metabolites of gliclizide (Gz). 7beta-hydroxy 123-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 17576805-0 2007 Characterization of diuron N-demethylation by mammalian hepatic microsomes and cDNA-expressed human cytochrome P450 enzymes. diuron n 20-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 17763085-2 2007 NDMA is acutely toxic to humans at high doses, is genotoxic after cytochrome P-450 metabolism, and is carcinogenic in several animal species. Dimethylnitrosamine 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 17823102-3 2007 PAHs are potent inducers of the hepatic cytochrome P-450 (CYP) isoenzymes 1A1, 1A2, and, possibly, 2E1. Polycyclic Aromatic Hydrocarbons 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 17823102-3 2007 PAHs are potent inducers of the hepatic cytochrome P-450 (CYP) isoenzymes 1A1, 1A2, and, possibly, 2E1. Polycyclic Aromatic Hydrocarbons 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 17676386-7 2007 Oral griseofulvin induced an increase in mitochondrial cytochrome P-450 levels, while chronic Isoflurane produced a reduction on its levels, without alterations on microsomal cytochrome P-450. Griseofulvin 5-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 17676386-8 2007 Allylisopropylacetamide diminished both mitochondrial and microsomal cytochrome P-450 brain levels; a similar pattern was detected in liver. Allylisopropylacetamide 0-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 17676386-9 2007 Mitochondria cytochrome P-450 liver levels were only diminished after chronic Isoflurane administration. Isoflurane 78-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 17676386-10 2007 In kidney only mitochondrial cytochrome P-450 levels were modified by veronal; while in microsomes, only acute anaesthesia with Enflurane diminished cytochrome P-450 content. Enflurane 128-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 17676386-12 2007 Acute delta-aminolevulinic acid administration reduced brain and liver cytochrome P-450 levels in both fractions; chronic delta-aminolevulinic acid administration diminished only liver mitochondrial cytochrome P-450. Aminolevulinic Acid 6-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 17676386-12 2007 Acute delta-aminolevulinic acid administration reduced brain and liver cytochrome P-450 levels in both fractions; chronic delta-aminolevulinic acid administration diminished only liver mitochondrial cytochrome P-450. Aminolevulinic Acid 122-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 199-215 17676386-13 2007 (5) Brain NADPH cytochrome P-450 reductase activity in animals receiving allylisopropylacetamide, dietary griseofulvin and delta-aminolevulinic acid showed a similar profile as that for total cytochrome P-450 levels. Allylisopropylacetamide 73-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 17676386-13 2007 (5) Brain NADPH cytochrome P-450 reductase activity in animals receiving allylisopropylacetamide, dietary griseofulvin and delta-aminolevulinic acid showed a similar profile as that for total cytochrome P-450 levels. Griseofulvin 106-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 17676386-13 2007 (5) Brain NADPH cytochrome P-450 reductase activity in animals receiving allylisopropylacetamide, dietary griseofulvin and delta-aminolevulinic acid showed a similar profile as that for total cytochrome P-450 levels. Aminolevulinic Acid 123-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 17434921-2 2007 Tamoxifen is metabolized by various cytochrome P450 (CYP) enzymes, but predominantly by CYP3A4. Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 17434921-2 2007 Tamoxifen is metabolized by various cytochrome P450 (CYP) enzymes, but predominantly by CYP3A4. Tamoxifen 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 17635335-0 2007 Effects of genetic polymorphism of cytochrome P450 enzymes on the pharmacokinetics of benzodiazepines. Benzodiazepines 86-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 17764757-1 2007 Cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) are lipid signalling molecules that elicit vasodilatation and modulate various intracellular signalling cascades. epoxyeicosatrienoic acids 30-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17764757-1 2007 Cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) are lipid signalling molecules that elicit vasodilatation and modulate various intracellular signalling cascades. epoxyeicosatrienoic acids 30-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 17764757-1 2007 Cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) are lipid signalling molecules that elicit vasodilatation and modulate various intracellular signalling cascades. eets 57-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17764757-1 2007 Cytochrome P450 (CYP)-derived epoxyeicosatrienoic acids (EETs) are lipid signalling molecules that elicit vasodilatation and modulate various intracellular signalling cascades. eets 57-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 26627620-1 2007 We have developed force-field parameters for the hydrogen-abstraction transition state of aliphatic hydroxylation by cytochrome P450 using the Q2MM approach. Hydrogen 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-132 17708140-5 2007 Interactions with warfarin, antidepressants, antiepileptic drugs, and statins often involve the cytochrome P450 enzymes. Warfarin 18-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-111 19356040-0 2007 cAMP-mediated regulation of CYP enzymes and its application in chemotherapy. Cyclic AMP 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 17635335-7 2007 Further studies on the effects of genetic polymorphisms of CYP enzymes on the pharmacokinetics and pharmacodynamics of BZPs are necessary to guide treatment individualization and optimization. Benzodiazepines 119-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 17505056-1 2007 P450 oxidoreductase (POR) has a pivotal role in facilitating electron transfer from nicotinamide adenine dinucleotide phosphate to microsomal cytochrome P450 (CYP) enzymes, including the steroidogenic enzymes CYP17A1 and CYP21A2. NADP 84-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 17654255-0 2007 Characterization of human liver cytochrome P-450 enzymes involved in the O-demethylation of a new P-glycoprotein inhibitor HM-30181. 4-oxo-4H-chromene-2-carboxylic acid (2-(2-(4-(2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl)-phenyl)-2H-tetrazol-5-yl)-4,5-dimethoxyphenyl)amide 123-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 4-oxo-4H-chromene-2-carboxylic acid (2-(2-(4-(2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl)-phenyl)-2H-tetrazol-5-yl)-4,5-dimethoxyphenyl)amide 108-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 4-oxo-4H-chromene-2-carboxylic acid (2-(2-(4-(2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl)-phenyl)-2H-tetrazol-5-yl)-4,5-dimethoxyphenyl)amide 108-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 4-oxo-4H-chromene-2-carboxylic acid (2-(2-(4-(2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl)-phenyl)-2H-tetrazol-5-yl)-4,5-dimethoxyphenyl)amide 108-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 251-254 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 4- or 5-o-desmethyl-hm-30181 120-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 4- or 5-o-desmethyl-hm-30181 120-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 6- or 7-o-desmethyl-hm-30181 158-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 17505056-1 2007 P450 oxidoreductase (POR) has a pivotal role in facilitating electron transfer from nicotinamide adenine dinucleotide phosphate to microsomal cytochrome P450 (CYP) enzymes, including the steroidogenic enzymes CYP17A1 and CYP21A2. NADP 84-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-162 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 6- or 7-o-desmethyl-hm-30181 158-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 17336049-4 2007 The potential of extracts of AP, AP formulations as well as HYP, its aglycone rooperol (ROP) and the sterols to inhibit in vitro metabolism of drug marker substrates by human cytochrome P450 (CYP) enzymes such as CYP3A4, 3A5 and CYP19 were investigated. CHEBI:166892 69-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-190 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 4-oxo-4H-chromene-2-carboxylic acid (2-(2-(4-(2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl)-phenyl)-2H-tetrazol-5-yl)-4,5-dimethoxyphenyl)amide 140-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 4-oxo-4H-chromene-2-carboxylic acid (2-(2-(4-(2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl)-phenyl)-2H-tetrazol-5-yl)-4,5-dimethoxyphenyl)amide 140-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 17654255-2 2007 This study was performed to characterize human cytochrome P-450 (CYP) enzymes involved in the metabolism of HM-30181 to 4- or 5-O-desmethyl-HM-30181 (M2) and 6- or 7-O-desmethyl-HM-30181 (M3) and to investigate the inhibitory potential of HM-30181 on CYP enzymes in human liver microsomes. 4-oxo-4H-chromene-2-carboxylic acid (2-(2-(4-(2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl)-phenyl)-2H-tetrazol-5-yl)-4,5-dimethoxyphenyl)amide 140-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 251-254 17654255-3 2007 CYP3A4 was identified as the major isozyme responsible for the O-demethylation of HM-30181 to M2 and M3 based on the correlation analysis, chemical inhibition and immuno-inhibition study and metabolism in cDNA-expressed human CYP isozymes. 4-oxo-4H-chromene-2-carboxylic acid (2-(2-(4-(2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)-ethyl)-phenyl)-2H-tetrazol-5-yl)-4,5-dimethoxyphenyl)amide 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 17336049-4 2007 The potential of extracts of AP, AP formulations as well as HYP, its aglycone rooperol (ROP) and the sterols to inhibit in vitro metabolism of drug marker substrates by human cytochrome P450 (CYP) enzymes such as CYP3A4, 3A5 and CYP19 were investigated. CHEBI:166892 69-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-195 17336049-4 2007 The potential of extracts of AP, AP formulations as well as HYP, its aglycone rooperol (ROP) and the sterols to inhibit in vitro metabolism of drug marker substrates by human cytochrome P450 (CYP) enzymes such as CYP3A4, 3A5 and CYP19 were investigated. rooperol 78-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-190 17336049-4 2007 The potential of extracts of AP, AP formulations as well as HYP, its aglycone rooperol (ROP) and the sterols to inhibit in vitro metabolism of drug marker substrates by human cytochrome P450 (CYP) enzymes such as CYP3A4, 3A5 and CYP19 were investigated. rooperol 78-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-195 17336049-4 2007 The potential of extracts of AP, AP formulations as well as HYP, its aglycone rooperol (ROP) and the sterols to inhibit in vitro metabolism of drug marker substrates by human cytochrome P450 (CYP) enzymes such as CYP3A4, 3A5 and CYP19 were investigated. Sterols 101-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-190 17336049-4 2007 The potential of extracts of AP, AP formulations as well as HYP, its aglycone rooperol (ROP) and the sterols to inhibit in vitro metabolism of drug marker substrates by human cytochrome P450 (CYP) enzymes such as CYP3A4, 3A5 and CYP19 were investigated. Sterols 101-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 192-195 17701830-3 2007 The specific CYPs responsible for O-demethylation of eupatilin to the major metabolite, 4-O-desmethyleupatilin were identified using a combination of correlation analysis, immuno-inhibition, chemical inhibition in human liver microsomes and metabolism by human cDNA-expressed CYP enzymes. 4-o-desmethyleupatilin 88-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 17413125-5 2007 One important xenobiotic substrate for CYP enzymes in cigarette smoke is polycyclic aromatic hydrocarbon, which in its native form is relatively harmless in small doses but upon bioactivation by CYP enzymes, can become very toxic substances for the lungs. Polycyclic Aromatic Hydrocarbons 73-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 17413125-5 2007 One important xenobiotic substrate for CYP enzymes in cigarette smoke is polycyclic aromatic hydrocarbon, which in its native form is relatively harmless in small doses but upon bioactivation by CYP enzymes, can become very toxic substances for the lungs. Polycyclic Aromatic Hydrocarbons 73-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-198 17638876-1 2007 Cytochrome P450 (CYP) epoxygenases convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EET), which exert diverse biological activities in a variety of systems. Arachidonic Acid 43-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17638876-1 2007 Cytochrome P450 (CYP) epoxygenases convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EET), which exert diverse biological activities in a variety of systems. Arachidonic Acid 43-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 17638876-1 2007 Cytochrome P450 (CYP) epoxygenases convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EET), which exert diverse biological activities in a variety of systems. epoxyeicosatrienoic acids 82-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17638876-1 2007 Cytochrome P450 (CYP) epoxygenases convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EET), which exert diverse biological activities in a variety of systems. epoxyeicosatrienoic acids 82-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 17638876-1 2007 Cytochrome P450 (CYP) epoxygenases convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EET), which exert diverse biological activities in a variety of systems. EET 109-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17638876-1 2007 Cytochrome P450 (CYP) epoxygenases convert arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EET), which exert diverse biological activities in a variety of systems. EET 109-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 17573638-1 2007 trans-Bromuconazole is a chiral chemical representative of a class of triazole derivatives known to inhibit specific fungal cytochrome P-450 (CYP) reactions. trans-bromuconazole 0-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 17573638-1 2007 trans-Bromuconazole is a chiral chemical representative of a class of triazole derivatives known to inhibit specific fungal cytochrome P-450 (CYP) reactions. trans-bromuconazole 0-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-145 17573638-1 2007 trans-Bromuconazole is a chiral chemical representative of a class of triazole derivatives known to inhibit specific fungal cytochrome P-450 (CYP) reactions. Triazoles 70-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 17573638-1 2007 trans-Bromuconazole is a chiral chemical representative of a class of triazole derivatives known to inhibit specific fungal cytochrome P-450 (CYP) reactions. Triazoles 70-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-145 17398093-0 2007 Imidazole moiety replacements in the 3-(1H-benzo[d]imidazol-2-yl)pyridin-2(1H)-one inhibitors of insulin-like growth factor receptor-1 (IGF-1R) to improve cytochrome P450 profile. 3-(1H-1,3-benzimidazol-2-yl)-2(1H)-pyridinone 37-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 17570247-0 2007 Cytochrome P450 isoforms catalyze formation of catechol estrogen quinones that react with DNA. catechol 47-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17570247-0 2007 Cytochrome P450 isoforms catalyze formation of catechol estrogen quinones that react with DNA. Quinones 65-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17497828-3 2007 The potential for inhibition of the CYP isoforms was measured by monitoring the level of the metabolites 6beta-hydroxytestosterone (3A4), dextrorphan (2D6), 4"-hydroxydiclofenac (2C9), and acetaminophen (1A2) formed in the presence of drug candidates using an eight-point titration. 6 beta-hydroxytestosterone 105-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 17452028-2 2007 Consequently dextromethorphan, flurbiprofen, midazolam and other compounds are commonly used as probe substrates to evaluate cytochrome P450 function in humans. Dextromethorphan 13-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-140 17452028-2 2007 Consequently dextromethorphan, flurbiprofen, midazolam and other compounds are commonly used as probe substrates to evaluate cytochrome P450 function in humans. Flurbiprofen 31-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-140 17452028-2 2007 Consequently dextromethorphan, flurbiprofen, midazolam and other compounds are commonly used as probe substrates to evaluate cytochrome P450 function in humans. Midazolam 45-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-140 17497828-3 2007 The potential for inhibition of the CYP isoforms was measured by monitoring the level of the metabolites 6beta-hydroxytestosterone (3A4), dextrorphan (2D6), 4"-hydroxydiclofenac (2C9), and acetaminophen (1A2) formed in the presence of drug candidates using an eight-point titration. Dextrorphan 138-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 17497828-3 2007 The potential for inhibition of the CYP isoforms was measured by monitoring the level of the metabolites 6beta-hydroxytestosterone (3A4), dextrorphan (2D6), 4"-hydroxydiclofenac (2C9), and acetaminophen (1A2) formed in the presence of drug candidates using an eight-point titration. 4'-hydroxydiclofenac 157-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 17497828-3 2007 The potential for inhibition of the CYP isoforms was measured by monitoring the level of the metabolites 6beta-hydroxytestosterone (3A4), dextrorphan (2D6), 4"-hydroxydiclofenac (2C9), and acetaminophen (1A2) formed in the presence of drug candidates using an eight-point titration. Acetaminophen 189-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 17398093-0 2007 Imidazole moiety replacements in the 3-(1H-benzo[d]imidazol-2-yl)pyridin-2(1H)-one inhibitors of insulin-like growth factor receptor-1 (IGF-1R) to improve cytochrome P450 profile. imidazole 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 17521300-0 2007 Artemisinin antimalarials moderately affect cytochrome P450 enzyme activity in healthy subjects. artemisinin 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 17521300-1 2007 The aim of this study was to investigate which principal human cytochrome P450 (CYP450) enzymes are affected by artemisinin and to what degree the artemisinin derivatives differ with respect to their respective induction and inhibition capacity. artemisinin 112-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 17521300-10 2007 These results show that intake of artemisinin antimalarials affect the activities of several principal human drug metabolizing CYP450 enzymes. artemisinin 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-133 17521300-1 2007 The aim of this study was to investigate which principal human cytochrome P450 (CYP450) enzymes are affected by artemisinin and to what degree the artemisinin derivatives differ with respect to their respective induction and inhibition capacity. artemisinin 112-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-86 17504220-0 2007 CYP-mediated clozapine interactions: how predictable are they? Clozapine 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 17449539-0 2007 Interaction between cytochrome P450 gene polymorphisms and serum organochlorine TEQ levels in the risk of endometriosis. organochlorine teq 65-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 17504219-1 2007 Cytochrome P450 (CYP, P450) is the collective term for a superfamily of heme-containing membrane proteins responsible for the metabolism of approximately 70 - 80 % of clinically used drugs. Heme 72-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17187958-0 2007 Cytochrome P450 destruction by benzene metabolites 1,4-benzoquinone and 1,4-hydroquinone and the formation of hydroxyl radicals in minipig liver microsomes. Benzene 31-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17187958-0 2007 Cytochrome P450 destruction by benzene metabolites 1,4-benzoquinone and 1,4-hydroquinone and the formation of hydroxyl radicals in minipig liver microsomes. quinone 51-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17187958-0 2007 Cytochrome P450 destruction by benzene metabolites 1,4-benzoquinone and 1,4-hydroquinone and the formation of hydroxyl radicals in minipig liver microsomes. 1,4-hydroquinone 72-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17187958-0 2007 Cytochrome P450 destruction by benzene metabolites 1,4-benzoquinone and 1,4-hydroquinone and the formation of hydroxyl radicals in minipig liver microsomes. Hydroxyl Radical 110-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17187958-2 2007 Since minipigs have been proposed as a suitable model species in toxicological and pharmacological research, the aim of this study was to explore mechanisms by which catechol, 1,4-hydroquinone and 1,4-benzoquinone destroy cytochrome P450 (P450) and induce oxidative stress in minipig liver microsomes. catechol 166-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-237 17187958-2 2007 Since minipigs have been proposed as a suitable model species in toxicological and pharmacological research, the aim of this study was to explore mechanisms by which catechol, 1,4-hydroquinone and 1,4-benzoquinone destroy cytochrome P450 (P450) and induce oxidative stress in minipig liver microsomes. 1,4-hydroquinone 176-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-237 17187958-2 2007 Since minipigs have been proposed as a suitable model species in toxicological and pharmacological research, the aim of this study was to explore mechanisms by which catechol, 1,4-hydroquinone and 1,4-benzoquinone destroy cytochrome P450 (P450) and induce oxidative stress in minipig liver microsomes. quinone 197-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-237 17504219-1 2007 Cytochrome P450 (CYP, P450) is the collective term for a superfamily of heme-containing membrane proteins responsible for the metabolism of approximately 70 - 80 % of clinically used drugs. Heme 72-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 17504220-4 2007 The majority of the interactions with clozapine are reported to be mediated by cytochrome P450 (CYP) enzymes. Clozapine 38-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 17504220-4 2007 The majority of the interactions with clozapine are reported to be mediated by cytochrome P450 (CYP) enzymes. Clozapine 38-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 17489643-5 2007 Of the drugs indicated for OAB, tolterodine, darifenacin, solifenacin, and oxybutynin are extensively metabolized by CYP450, but trospium is not. Tolterodine Tartrate 32-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-123 17489643-5 2007 Of the drugs indicated for OAB, tolterodine, darifenacin, solifenacin, and oxybutynin are extensively metabolized by CYP450, but trospium is not. darifenacin 45-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-123 17489643-5 2007 Of the drugs indicated for OAB, tolterodine, darifenacin, solifenacin, and oxybutynin are extensively metabolized by CYP450, but trospium is not. Solifenacin Succinate 58-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-123 17489643-5 2007 Of the drugs indicated for OAB, tolterodine, darifenacin, solifenacin, and oxybutynin are extensively metabolized by CYP450, but trospium is not. oxybutynin 75-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-123 17372695-0 2007 Novel cyclic phosphate prodrug approach for cytochrome P450-activated drugs containing an alcohol functionality. cyclic phosphate 6-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 17293117-0 2007 Cytochrome P-450 model reactions: efficient and highly selective oxidation of alcohols with tetrabutylammonium peroxymonosulfate catalyzed by Mn-porphyrins. Alcohols 78-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 17293117-0 2007 Cytochrome P-450 model reactions: efficient and highly selective oxidation of alcohols with tetrabutylammonium peroxymonosulfate catalyzed by Mn-porphyrins. SCHEMBL1259012 92-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 17293117-0 2007 Cytochrome P-450 model reactions: efficient and highly selective oxidation of alcohols with tetrabutylammonium peroxymonosulfate catalyzed by Mn-porphyrins. mn-porphyrins 142-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 17220239-11 2007 CYP3A4 was the major cytochrome P450 isozyme responsible for the limited oxidative metabolism of sitagliptin, with some minor contribution from CYP2C8. Sitagliptin Phosphate 97-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 17381666-0 2007 Rifampin, a cytochrome P450 3A inducer, decreases plasma concentrations of antipsychotic risperidone in healthy volunteers. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 17381666-1 2007 BACKGROUND: Although cytochrome P450 (CYP) 2D6 is often thought to be the only CYP responsible for the metabolism of risperidone, many reports suggest that CYP3A may be involved too. Risperidone 117-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 17381666-1 2007 BACKGROUND: Although cytochrome P450 (CYP) 2D6 is often thought to be the only CYP responsible for the metabolism of risperidone, many reports suggest that CYP3A may be involved too. Risperidone 117-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 17381666-1 2007 BACKGROUND: Although cytochrome P450 (CYP) 2D6 is often thought to be the only CYP responsible for the metabolism of risperidone, many reports suggest that CYP3A may be involved too. Risperidone 117-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 17407229-0 2007 Cytochrome P450 polymorphism as a predictor of ovarian toxicity to pulse cyclophosphamide in systemic lupus erythematosus. Cyclophosphamide 73-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17372695-0 2007 Novel cyclic phosphate prodrug approach for cytochrome P450-activated drugs containing an alcohol functionality. Alcohols 90-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 17372695-1 2007 PURPOSE: A cyclic phosphate prodrug of a descriptive molecule containing an alcohol functionality was designed, synthesized and characterized in vitro as a cytochrome P450 (CYP) -selective prodrug. cyclic phosphate 11-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-171 17372695-1 2007 PURPOSE: A cyclic phosphate prodrug of a descriptive molecule containing an alcohol functionality was designed, synthesized and characterized in vitro as a cytochrome P450 (CYP) -selective prodrug. cyclic phosphate 11-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-176 17372695-1 2007 PURPOSE: A cyclic phosphate prodrug of a descriptive molecule containing an alcohol functionality was designed, synthesized and characterized in vitro as a cytochrome P450 (CYP) -selective prodrug. Alcohols 76-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-171 17372695-1 2007 PURPOSE: A cyclic phosphate prodrug of a descriptive molecule containing an alcohol functionality was designed, synthesized and characterized in vitro as a cytochrome P450 (CYP) -selective prodrug. Alcohols 76-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-176 17372695-2 2007 MATERIALS AND METHODS: To achieve efficient CYP-oxidation and prodrug bioconversion, 1,3-cyclic propyl ester of phosphate was designed to have a C4-aryl substituent and synthesized using phosphorus(III) chemistry. 1,3-cyclic propyl ester 85-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 17372695-2 2007 MATERIALS AND METHODS: To achieve efficient CYP-oxidation and prodrug bioconversion, 1,3-cyclic propyl ester of phosphate was designed to have a C4-aryl substituent and synthesized using phosphorus(III) chemistry. Phosphates 112-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 17372695-3 2007 The two-step bioconversion of the cyclic phosphate prodrug was evaluated in vitro using human liver microsomes and recombinant CYP enzymes. cyclic phosphate 34-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 17372695-4 2007 RESULTS: This cyclic phosphate prodrug underwent initial CYP-catalyzed oxidation and was mainly catalyzed by the CYP3A4 form. cyclic phosphate 14-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 17372695-8 2007 CONCLUSIONS: Since CYP enzymes reside predominantly in the liver and secondarily in the small intestine, the results indicate that cyclic phosphate prodrugs represent a very feasible liver- or intestinal-targeted drug delivery strategy for drug molecules containing an alcohol functionality. cyclic phosphate 131-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 17372695-8 2007 CONCLUSIONS: Since CYP enzymes reside predominantly in the liver and secondarily in the small intestine, the results indicate that cyclic phosphate prodrugs represent a very feasible liver- or intestinal-targeted drug delivery strategy for drug molecules containing an alcohol functionality. Alcohols 269-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 17303175-0 2007 Cytochrome P450 enzymes involved in the metabolism of tetrahydrocannabinols and cannabinol by human hepatic microsomes. Dronabinol 54-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17292855-0 2007 Evaluation of cytochrome P450(BSbeta) reactivity against polycyclic aromatic hydrocarbons and drugs. Polycyclic Aromatic Hydrocarbons 57-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-37 17292855-1 2007 The oxidation of 10 polycyclic aromatic hydrocarbons (PAH) by cytochrome P450(BSbeta) using three different electron acceptors is reported. Polycyclic Aromatic Hydrocarbons 20-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-85 17292855-1 2007 The oxidation of 10 polycyclic aromatic hydrocarbons (PAH) by cytochrome P450(BSbeta) using three different electron acceptors is reported. Polycyclic Aromatic Hydrocarbons 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-85 17303175-0 2007 Cytochrome P450 enzymes involved in the metabolism of tetrahydrocannabinols and cannabinol by human hepatic microsomes. Cannabinol 64-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17160483-0 2007 Metabolism of N-methyl, N-propargylphenylethylamine: studies with human liver microsomes and cDNA expressed cytochrome P450 (CYP) enzymes. n-methyl, n-propargylphenylethylamine 14-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-123 17322161-4 2007 Cranberry juice is a flavonoid, which has been shown to induce, inhibit, or act as a substrate for the biosynthesis of several cytochrome P-450 (CYP) isoenzymes. Flavonoids 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-143 17322161-4 2007 Cranberry juice is a flavonoid, which has been shown to induce, inhibit, or act as a substrate for the biosynthesis of several cytochrome P-450 (CYP) isoenzymes. Flavonoids 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 17160483-0 2007 Metabolism of N-methyl, N-propargylphenylethylamine: studies with human liver microsomes and cDNA expressed cytochrome P450 (CYP) enzymes. n-methyl, n-propargylphenylethylamine 14-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 17160483-2 2007 We used an in vitro screening procedure and studies with individual human liver microsomes and cDNA-expressed CYP enzymes to investigate the metabolism of the putative neuroprotective drug N-methyl,N-propargyl-2-phenylethylamine (MPPE) to N-methylphenylethylamine (N-methylPEA) and N-propargylphenylethylamine (N-propargylPEA). n-methyl,n-propargyl-2-phenylethylamine 189-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 17160483-2 2007 We used an in vitro screening procedure and studies with individual human liver microsomes and cDNA-expressed CYP enzymes to investigate the metabolism of the putative neuroprotective drug N-methyl,N-propargyl-2-phenylethylamine (MPPE) to N-methylphenylethylamine (N-methylPEA) and N-propargylphenylethylamine (N-propargylPEA). MPPE 230-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 17160483-2 2007 We used an in vitro screening procedure and studies with individual human liver microsomes and cDNA-expressed CYP enzymes to investigate the metabolism of the putative neuroprotective drug N-methyl,N-propargyl-2-phenylethylamine (MPPE) to N-methylphenylethylamine (N-methylPEA) and N-propargylphenylethylamine (N-propargylPEA). N-methylphenethylamine 239-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 17160483-2 2007 We used an in vitro screening procedure and studies with individual human liver microsomes and cDNA-expressed CYP enzymes to investigate the metabolism of the putative neuroprotective drug N-methyl,N-propargyl-2-phenylethylamine (MPPE) to N-methylphenylethylamine (N-methylPEA) and N-propargylphenylethylamine (N-propargylPEA). N-methylphenethylamine 265-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 17160483-2 2007 We used an in vitro screening procedure and studies with individual human liver microsomes and cDNA-expressed CYP enzymes to investigate the metabolism of the putative neuroprotective drug N-methyl,N-propargyl-2-phenylethylamine (MPPE) to N-methylphenylethylamine (N-methylPEA) and N-propargylphenylethylamine (N-propargylPEA). n-propargylphenylethylamine 282-309 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 17160483-2 2007 We used an in vitro screening procedure and studies with individual human liver microsomes and cDNA-expressed CYP enzymes to investigate the metabolism of the putative neuroprotective drug N-methyl,N-propargyl-2-phenylethylamine (MPPE) to N-methylphenylethylamine (N-methylPEA) and N-propargylphenylethylamine (N-propargylPEA). n-propargylpea 311-325 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 17329906-0 2007 Cytochrome P450 is responsible for nitric oxide generation from NO-aspirin and other organic nitrates. Nitric Oxide 35-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17346171-1 2007 This article aims to give an overview on the characterization, properties and regulation of enzymes, particularly the cytochrome (CYP) P450 enzymes, in the formation of bile acids from cholesterol. Bile Acids and Salts 169-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 17346171-1 2007 This article aims to give an overview on the characterization, properties and regulation of enzymes, particularly the cytochrome (CYP) P450 enzymes, in the formation of bile acids from cholesterol. Cholesterol 185-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 17237483-1 2007 Pro-carcinogens, such as benzo[a]pyrene (B[a]P), that are exogenous ligands of the aromatic hydrocarbon receptor may influence the susceptibility of target-cell populations through the up-regulation of cytochrome P450 (CYP) mixed function oxidases. Benzo(a)pyrene 25-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-217 17237483-1 2007 Pro-carcinogens, such as benzo[a]pyrene (B[a]P), that are exogenous ligands of the aromatic hydrocarbon receptor may influence the susceptibility of target-cell populations through the up-regulation of cytochrome P450 (CYP) mixed function oxidases. Benzo(a)pyrene 25-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 219-222 17237483-1 2007 Pro-carcinogens, such as benzo[a]pyrene (B[a]P), that are exogenous ligands of the aromatic hydrocarbon receptor may influence the susceptibility of target-cell populations through the up-regulation of cytochrome P450 (CYP) mixed function oxidases. Benzo(a)pyrene 41-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-217 17237483-1 2007 Pro-carcinogens, such as benzo[a]pyrene (B[a]P), that are exogenous ligands of the aromatic hydrocarbon receptor may influence the susceptibility of target-cell populations through the up-regulation of cytochrome P450 (CYP) mixed function oxidases. Benzo(a)pyrene 41-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 219-222 17329906-0 2007 Cytochrome P450 is responsible for nitric oxide generation from NO-aspirin and other organic nitrates. Aspirin 67-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17329906-0 2007 Cytochrome P450 is responsible for nitric oxide generation from NO-aspirin and other organic nitrates. Nitrates 93-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17329906-2 2007 We have previously reported that cytochrome P450 (P450) plays important role in NO generation from other organic nitrates such as nitroglycerin (NTG) and isosorbide dinitrate (ISDN). Nitrates 113-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 17329906-2 2007 We have previously reported that cytochrome P450 (P450) plays important role in NO generation from other organic nitrates such as nitroglycerin (NTG) and isosorbide dinitrate (ISDN). Nitroglycerin 130-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 17329906-2 2007 We have previously reported that cytochrome P450 (P450) plays important role in NO generation from other organic nitrates such as nitroglycerin (NTG) and isosorbide dinitrate (ISDN). Nitroglycerin 145-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 17329906-2 2007 We have previously reported that cytochrome P450 (P450) plays important role in NO generation from other organic nitrates such as nitroglycerin (NTG) and isosorbide dinitrate (ISDN). Isosorbide Dinitrate 154-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 17329906-2 2007 We have previously reported that cytochrome P450 (P450) plays important role in NO generation from other organic nitrates such as nitroglycerin (NTG) and isosorbide dinitrate (ISDN). Isosorbide Dinitrate 176-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 17222962-1 2007 Cytochrome P450 side-chain cleavage (CYP11A1) catalyzes the conversion of cholesterol to pregnenolone, the first step in steroidogenesis. Cholesterol 74-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17253801-0 2007 Regioselective oxyfunctionalization of unactivated carbons in steroids by a model of cytochrome P-450: osmiumporphyrin complex/tert-butyl hydroperoxide system. Carbon 51-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 17253801-0 2007 Regioselective oxyfunctionalization of unactivated carbons in steroids by a model of cytochrome P-450: osmiumporphyrin complex/tert-butyl hydroperoxide system. Steroids 62-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 17253801-0 2007 Regioselective oxyfunctionalization of unactivated carbons in steroids by a model of cytochrome P-450: osmiumporphyrin complex/tert-butyl hydroperoxide system. osmiumporphyrin complex 103-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 17253801-0 2007 Regioselective oxyfunctionalization of unactivated carbons in steroids by a model of cytochrome P-450: osmiumporphyrin complex/tert-butyl hydroperoxide system. tert-Butylhydroperoxide 127-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 17222962-1 2007 Cytochrome P450 side-chain cleavage (CYP11A1) catalyzes the conversion of cholesterol to pregnenolone, the first step in steroidogenesis. Pregnenolone 89-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 17484517-0 2007 Drug-drug interactions in the metabolism of imidafenacin: role of the human cytochrome P450 enzymes and UDP-glucuronic acid transferases, and potential of imidafenacin to inhibit human cytochrome P450 enzymes. imidafenacin 44-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 17286541-5 2007 Amodiaquine is almost entirely metabolized by the polymorphic cytochrome P450 (CYP) isoform 2C8 to the pharmacologically active desethylamodiaquine. Amodiaquine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 17286541-5 2007 Amodiaquine is almost entirely metabolized by the polymorphic cytochrome P450 (CYP) isoform 2C8 to the pharmacologically active desethylamodiaquine. Amodiaquine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 17286541-5 2007 Amodiaquine is almost entirely metabolized by the polymorphic cytochrome P450 (CYP) isoform 2C8 to the pharmacologically active desethylamodiaquine. desethylamodiaquine 128-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 17286541-5 2007 Amodiaquine is almost entirely metabolized by the polymorphic cytochrome P450 (CYP) isoform 2C8 to the pharmacologically active desethylamodiaquine. desethylamodiaquine 128-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 17484517-0 2007 Drug-drug interactions in the metabolism of imidafenacin: role of the human cytochrome P450 enzymes and UDP-glucuronic acid transferases, and potential of imidafenacin to inhibit human cytochrome P450 enzymes. imidafenacin 155-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-200 17484519-0 2007 Identification of human cytochrome P450 enzymes involved in the major metabolic pathway of fluvoxamine. Fluvoxamine 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 17484519-2 2007 The present study was carried out to identify the cytochrome P450 (CYP) enzyme(s) involved in the metabolism offluvoxamine to fluvoxamino alcohol using human liver microsomes and cDNA-expressed human CYP enzymes. offluvoxamine 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 17484519-2 2007 The present study was carried out to identify the cytochrome P450 (CYP) enzyme(s) involved in the metabolism offluvoxamine to fluvoxamino alcohol using human liver microsomes and cDNA-expressed human CYP enzymes. offluvoxamine 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 17484519-2 2007 The present study was carried out to identify the cytochrome P450 (CYP) enzyme(s) involved in the metabolism offluvoxamine to fluvoxamino alcohol using human liver microsomes and cDNA-expressed human CYP enzymes. offluvoxamine 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-203 17484519-2 2007 The present study was carried out to identify the cytochrome P450 (CYP) enzyme(s) involved in the metabolism offluvoxamine to fluvoxamino alcohol using human liver microsomes and cDNA-expressed human CYP enzymes. fluvoxamino alcohol 126-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 17484519-2 2007 The present study was carried out to identify the cytochrome P450 (CYP) enzyme(s) involved in the metabolism offluvoxamine to fluvoxamino alcohol using human liver microsomes and cDNA-expressed human CYP enzymes. fluvoxamino alcohol 126-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 17484519-2 2007 The present study was carried out to identify the cytochrome P450 (CYP) enzyme(s) involved in the metabolism offluvoxamine to fluvoxamino alcohol using human liver microsomes and cDNA-expressed human CYP enzymes. fluvoxamino alcohol 126-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-203 17484519-4 2007 The formation of fluvoxamino alcohol from fluvoxamine in pooled human liver microsomes was significantly inhibited by quinidine, a relatively specific CYP2D6 inhibitor, with a Ki value of 2.2 microM, whereas other several relatively specific CYP inhibitors did not inhibit the formation of fluvoxamino alcohol. fluvoxamino alcohol 17-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 17484519-4 2007 The formation of fluvoxamino alcohol from fluvoxamine in pooled human liver microsomes was significantly inhibited by quinidine, a relatively specific CYP2D6 inhibitor, with a Ki value of 2.2 microM, whereas other several relatively specific CYP inhibitors did not inhibit the formation of fluvoxamino alcohol. Fluvoxamine 42-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 17484519-4 2007 The formation of fluvoxamino alcohol from fluvoxamine in pooled human liver microsomes was significantly inhibited by quinidine, a relatively specific CYP2D6 inhibitor, with a Ki value of 2.2 microM, whereas other several relatively specific CYP inhibitors did not inhibit the formation of fluvoxamino alcohol. Quinidine 118-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 17484519-5 2007 In addition, only CYP2D6 of several cDNA-expressed human CYP enzymes examined showed substantial activity for the formation of fluvoxamino alcohol. fluvoxamino alcohol 127-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 17067556-0 2007 Identification of monoamine oxidase and cytochrome P450 isoenzymes involved in the deamination of phenethylamine-derived designer drugs (2C-series). phenethylamine 98-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 17201456-5 2007 Because they are metabolised via cytochrome P450 (CYP), glitazones are exposed to numerous pharmacokinetic interactions. Thiazolidinediones 56-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 17082249-1 2007 BACKGROUND: CYP2B6 is a highly variable and polymorphic cytochrome P450 (CYP) enzyme involved in the biotransformation of an increasing number of drugs, including cyclophosphamide, bupropion, and the nonnucleosidic reverse transcriptase inhibitor efavirenz. Cyclophosphamide 163-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 17082249-1 2007 BACKGROUND: CYP2B6 is a highly variable and polymorphic cytochrome P450 (CYP) enzyme involved in the biotransformation of an increasing number of drugs, including cyclophosphamide, bupropion, and the nonnucleosidic reverse transcriptase inhibitor efavirenz. Cyclophosphamide 163-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 17082249-1 2007 BACKGROUND: CYP2B6 is a highly variable and polymorphic cytochrome P450 (CYP) enzyme involved in the biotransformation of an increasing number of drugs, including cyclophosphamide, bupropion, and the nonnucleosidic reverse transcriptase inhibitor efavirenz. Bupropion 181-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 17082249-1 2007 BACKGROUND: CYP2B6 is a highly variable and polymorphic cytochrome P450 (CYP) enzyme involved in the biotransformation of an increasing number of drugs, including cyclophosphamide, bupropion, and the nonnucleosidic reverse transcriptase inhibitor efavirenz. Bupropion 181-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 17082249-1 2007 BACKGROUND: CYP2B6 is a highly variable and polymorphic cytochrome P450 (CYP) enzyme involved in the biotransformation of an increasing number of drugs, including cyclophosphamide, bupropion, and the nonnucleosidic reverse transcriptase inhibitor efavirenz. efavirenz 247-256 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 17082249-1 2007 BACKGROUND: CYP2B6 is a highly variable and polymorphic cytochrome P450 (CYP) enzyme involved in the biotransformation of an increasing number of drugs, including cyclophosphamide, bupropion, and the nonnucleosidic reverse transcriptase inhibitor efavirenz. efavirenz 247-256 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 17375980-14 2007 Escitalopram is metabolised by the cytochrome P450 (CYP) isoenzymes CYP2C19, CYP2D6 and CYP3A4. Citalopram 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 17375980-14 2007 Escitalopram is metabolised by the cytochrome P450 (CYP) isoenzymes CYP2C19, CYP2D6 and CYP3A4. Citalopram 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 17375983-7 2007 The body surface area, peak concentrations of etoposide, urinary recovery of dechloroethylcyclophosphamide (formed by cytochrome P450 [CYP] 3A4) relative to the cyclophosphamide dose and number of cycles had a significant effect on toxicity. dechloroethylcyclophosphamide 77-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-143 17375983-7 2007 The body surface area, peak concentrations of etoposide, urinary recovery of dechloroethylcyclophosphamide (formed by cytochrome P450 [CYP] 3A4) relative to the cyclophosphamide dose and number of cycles had a significant effect on toxicity. Cyclophosphamide 90-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-143 17201456-5 2007 Because they are metabolised via cytochrome P450 (CYP), glitazones are exposed to numerous pharmacokinetic interactions. Thiazolidinediones 56-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 17201456-11 2007 Although some food components have also been shown to potentially interfere with drugs metabolised with the CYP system, no published study deals specifically with these possible CYP-mediated food-drug interactions with glitazones. Thiazolidinediones 219-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 178-181 19356015-2 2007 The CYP form with the lowest in vitro K(i) is evaluated first clinically, employing a suitable probe drug like midazolam (CYP3A4), theophylline (CYP1A2), (S)-warfarin (CYP2C9) and desipramine (CYP2D6), and the NCE is classified as a "none", "weak", "moderate", or "strong" inhibitor. Midazolam 111-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 18690854-16 2007 These interactions are caused by the metabolism of triazoles in the liver where the cytochrome P450 (CYP) system is involved at a different extend as well as by their mechanisms of action. Triazoles 51-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 18690854-16 2007 These interactions are caused by the metabolism of triazoles in the liver where the cytochrome P450 (CYP) system is involved at a different extend as well as by their mechanisms of action. Triazoles 51-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 18447001-0 2007 The effect of chlorpyrifos-oxon and other xenobiotics on the human cytochrome P450-dependent metabolism of naphthalene and deet. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 14-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 18447001-0 2007 The effect of chlorpyrifos-oxon and other xenobiotics on the human cytochrome P450-dependent metabolism of naphthalene and deet. naphthalene 107-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 17674211-5 2007 To explain the metabolic dephenylation process, it has been suggested that PBNA is metabolized by cytochrome P-450 (CYP) enzymes to the phenolic derivative 4"-hydroxy-N-phenyl-2-naphthylamine, followed by its further oxidation to the quinone imine, which subsequently hydrolyses to form the dephenylation product 2-naphthylamine. neozone 75-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 17674211-5 2007 To explain the metabolic dephenylation process, it has been suggested that PBNA is metabolized by cytochrome P-450 (CYP) enzymes to the phenolic derivative 4"-hydroxy-N-phenyl-2-naphthylamine, followed by its further oxidation to the quinone imine, which subsequently hydrolyses to form the dephenylation product 2-naphthylamine. neozone 75-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 17674211-5 2007 To explain the metabolic dephenylation process, it has been suggested that PBNA is metabolized by cytochrome P-450 (CYP) enzymes to the phenolic derivative 4"-hydroxy-N-phenyl-2-naphthylamine, followed by its further oxidation to the quinone imine, which subsequently hydrolyses to form the dephenylation product 2-naphthylamine. 4'-hydroxy-n-phenyl-2-naphthylamine 156-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 17674211-5 2007 To explain the metabolic dephenylation process, it has been suggested that PBNA is metabolized by cytochrome P-450 (CYP) enzymes to the phenolic derivative 4"-hydroxy-N-phenyl-2-naphthylamine, followed by its further oxidation to the quinone imine, which subsequently hydrolyses to form the dephenylation product 2-naphthylamine. 4'-hydroxy-n-phenyl-2-naphthylamine 156-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 17674211-5 2007 To explain the metabolic dephenylation process, it has been suggested that PBNA is metabolized by cytochrome P-450 (CYP) enzymes to the phenolic derivative 4"-hydroxy-N-phenyl-2-naphthylamine, followed by its further oxidation to the quinone imine, which subsequently hydrolyses to form the dephenylation product 2-naphthylamine. quinone imine 234-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 17674211-5 2007 To explain the metabolic dephenylation process, it has been suggested that PBNA is metabolized by cytochrome P-450 (CYP) enzymes to the phenolic derivative 4"-hydroxy-N-phenyl-2-naphthylamine, followed by its further oxidation to the quinone imine, which subsequently hydrolyses to form the dephenylation product 2-naphthylamine. quinone imine 234-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 17674211-5 2007 To explain the metabolic dephenylation process, it has been suggested that PBNA is metabolized by cytochrome P-450 (CYP) enzymes to the phenolic derivative 4"-hydroxy-N-phenyl-2-naphthylamine, followed by its further oxidation to the quinone imine, which subsequently hydrolyses to form the dephenylation product 2-naphthylamine. 2-Naphthylamine 176-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 17674211-5 2007 To explain the metabolic dephenylation process, it has been suggested that PBNA is metabolized by cytochrome P-450 (CYP) enzymes to the phenolic derivative 4"-hydroxy-N-phenyl-2-naphthylamine, followed by its further oxidation to the quinone imine, which subsequently hydrolyses to form the dephenylation product 2-naphthylamine. 2-Naphthylamine 176-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 17786620-1 2007 Ronald Estabrook made his initial impact studying cytochrome P450 by demonstrating the oxidative metabolism function of this unique class of enzymes, which had an unusual spectral peak at 450 nm when reduced and in the presence of carbon monoxide. Carbon Monoxide 231-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 17786620-2 2007 Utilizing a photochemical action spectrum, he demonstrated that a cytochrome P450 was responsible for steroid 21 hydroxylation catalyzed by microsomes prepared from adrenal cortex tissue. Steroids 102-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 19356015-2 2007 The CYP form with the lowest in vitro K(i) is evaluated first clinically, employing a suitable probe drug like midazolam (CYP3A4), theophylline (CYP1A2), (S)-warfarin (CYP2C9) and desipramine (CYP2D6), and the NCE is classified as a "none", "weak", "moderate", or "strong" inhibitor. Theophylline 131-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 19356015-2 2007 The CYP form with the lowest in vitro K(i) is evaluated first clinically, employing a suitable probe drug like midazolam (CYP3A4), theophylline (CYP1A2), (S)-warfarin (CYP2C9) and desipramine (CYP2D6), and the NCE is classified as a "none", "weak", "moderate", or "strong" inhibitor. Warfarin 156-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 19356015-2 2007 The CYP form with the lowest in vitro K(i) is evaluated first clinically, employing a suitable probe drug like midazolam (CYP3A4), theophylline (CYP1A2), (S)-warfarin (CYP2C9) and desipramine (CYP2D6), and the NCE is classified as a "none", "weak", "moderate", or "strong" inhibitor. Desipramine 180-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 17936932-6 2007 A number of pesticides, including chlorpyrifos, fipronil and permethrin, and the repellent, DEET, have been shown to be inducers of CYP isoforms in human hepatocytes, with fipronil being the most potent. Chlorpyrifos 34-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 17427179-0 2007 Inhibition of fipronil and nonane metabolism in human liver microsomes and human cytochrome P450 isoforms by chlorpyrifos. fipronil 14-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 17936932-6 2007 A number of pesticides, including chlorpyrifos, fipronil and permethrin, and the repellent, DEET, have been shown to be inducers of CYP isoforms in human hepatocytes, with fipronil being the most potent. fipronil 48-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 17427179-0 2007 Inhibition of fipronil and nonane metabolism in human liver microsomes and human cytochrome P450 isoforms by chlorpyrifos. nonane 27-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 17936932-3 2007 OPs are potent irreversible inhibitors of testosterone metabolism by cytochrome P450 (CYP) 3A4 and of estradiol metabolism by CYP3A4 and CYP1A2. Testosterone 42-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 17936932-6 2007 A number of pesticides, including chlorpyrifos, fipronil and permethrin, and the repellent, DEET, have been shown to be inducers of CYP isoforms in human hepatocytes, with fipronil being the most potent. Permethrin 61-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 17936932-3 2007 OPs are potent irreversible inhibitors of testosterone metabolism by cytochrome P450 (CYP) 3A4 and of estradiol metabolism by CYP3A4 and CYP1A2. Testosterone 42-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 17936932-4 2007 All of these CYP inhibitions are believed to be due to the release of reactive sulfur during CYP-catalyzed oxidative desulfuration. Sulfur 79-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 17427179-0 2007 Inhibition of fipronil and nonane metabolism in human liver microsomes and human cytochrome P450 isoforms by chlorpyrifos. Chlorpyrifos 109-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 17427179-1 2007 Previous studies have established that chlorpyrifos (CPS), fipronil, and nonane can all be metabolized by human liver microsomes (HLM) and a number of cytochrome P450 (CYP) isoforms. Chlorpyrifos 39-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-166 17427179-1 2007 Previous studies have established that chlorpyrifos (CPS), fipronil, and nonane can all be metabolized by human liver microsomes (HLM) and a number of cytochrome P450 (CYP) isoforms. Chlorpyrifos 39-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-171 17936932-6 2007 A number of pesticides, including chlorpyrifos, fipronil and permethrin, and the repellent, DEET, have been shown to be inducers of CYP isoforms in human hepatocytes, with fipronil being the most potent. fipronil 172-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 17427179-1 2007 Previous studies have established that chlorpyrifos (CPS), fipronil, and nonane can all be metabolized by human liver microsomes (HLM) and a number of cytochrome P450 (CYP) isoforms. Chlorpyrifos 53-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-166 17936932-4 2007 All of these CYP inhibitions are believed to be due to the release of reactive sulfur during CYP-catalyzed oxidative desulfuration. Sulfur 79-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 17164129-1 2007 Arachidonic acid (AA) is an essential fatty acid that is metabolized by cyclooxygenase (COX), lipoxygenase (LOX) or cytochrome P450 (CYP) enzymes to generate eicosanoids which in turn mediate a number of biological activities including regulation of angiogenesis. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 17011724-0 2007 Significant transcriptional down-regulation of the human MDR1 gene by beta-naphthoflavone: a proposed hypothesis linking potent CYP gene induction to MDR1 inhibition. beta-Naphthoflavone 70-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 17507873-1 2007 Isoflavonoids exert a regulatory function on the expression of cytochrome P450 enzymes and also up-regulate the vitamin D(3) receptor (VDR) on cancer cells, which increase their sensitivity to 1,25-dihydroxyvitamin D(3) , the hormonally active form of vitamin D(3) . isoflavonoids 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 17427179-1 2007 Previous studies have established that chlorpyrifos (CPS), fipronil, and nonane can all be metabolized by human liver microsomes (HLM) and a number of cytochrome P450 (CYP) isoforms. Chlorpyrifos 53-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-171 17427179-1 2007 Previous studies have established that chlorpyrifos (CPS), fipronil, and nonane can all be metabolized by human liver microsomes (HLM) and a number of cytochrome P450 (CYP) isoforms. fipronil 59-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-166 17427179-1 2007 Previous studies have established that chlorpyrifos (CPS), fipronil, and nonane can all be metabolized by human liver microsomes (HLM) and a number of cytochrome P450 (CYP) isoforms. fipronil 59-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-171 17427179-1 2007 Previous studies have established that chlorpyrifos (CPS), fipronil, and nonane can all be metabolized by human liver microsomes (HLM) and a number of cytochrome P450 (CYP) isoforms. nonane 73-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-166 17427179-1 2007 Previous studies have established that chlorpyrifos (CPS), fipronil, and nonane can all be metabolized by human liver microsomes (HLM) and a number of cytochrome P450 (CYP) isoforms. nonane 73-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-171 17427179-3 2007 In this study the effect of either coincubation or preincubation of CPS with HLM or CYP isoforms with either fipronil or nonane as substrate was investigated. fipronil 109-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 17427179-3 2007 In this study the effect of either coincubation or preincubation of CPS with HLM or CYP isoforms with either fipronil or nonane as substrate was investigated. nonane 121-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 17164129-0 2007 Emerging mechanisms for growth and protection of the vasculature by cytochrome P450-derived products of arachidonic acid and other eicosanoids. Arachidonic Acid 104-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 17164129-0 2007 Emerging mechanisms for growth and protection of the vasculature by cytochrome P450-derived products of arachidonic acid and other eicosanoids. Eicosanoids 131-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 17164129-1 2007 Arachidonic acid (AA) is an essential fatty acid that is metabolized by cyclooxygenase (COX), lipoxygenase (LOX) or cytochrome P450 (CYP) enzymes to generate eicosanoids which in turn mediate a number of biological activities including regulation of angiogenesis. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 17164129-1 2007 Arachidonic acid (AA) is an essential fatty acid that is metabolized by cyclooxygenase (COX), lipoxygenase (LOX) or cytochrome P450 (CYP) enzymes to generate eicosanoids which in turn mediate a number of biological activities including regulation of angiogenesis. Fatty Acids, Essential 28-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 17164129-1 2007 Arachidonic acid (AA) is an essential fatty acid that is metabolized by cyclooxygenase (COX), lipoxygenase (LOX) or cytochrome P450 (CYP) enzymes to generate eicosanoids which in turn mediate a number of biological activities including regulation of angiogenesis. Fatty Acids, Essential 28-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 17164129-1 2007 Arachidonic acid (AA) is an essential fatty acid that is metabolized by cyclooxygenase (COX), lipoxygenase (LOX) or cytochrome P450 (CYP) enzymes to generate eicosanoids which in turn mediate a number of biological activities including regulation of angiogenesis. Eicosanoids 158-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 17164129-1 2007 Arachidonic acid (AA) is an essential fatty acid that is metabolized by cyclooxygenase (COX), lipoxygenase (LOX) or cytochrome P450 (CYP) enzymes to generate eicosanoids which in turn mediate a number of biological activities including regulation of angiogenesis. Eicosanoids 158-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 17164129-4 2007 A number of these enzymes, including members of the CYP 4 family, predominantly catalyze conversion of AA to 20-hydroxyeicosatetraenoic acid (20-HETE) while the CYP epoxygenases generate mainly epoxyeicosatrienoic acids (EETs). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 109-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 17164129-4 2007 A number of these enzymes, including members of the CYP 4 family, predominantly catalyze conversion of AA to 20-hydroxyeicosatetraenoic acid (20-HETE) while the CYP epoxygenases generate mainly epoxyeicosatrienoic acids (EETs). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 142-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 17164129-4 2007 A number of these enzymes, including members of the CYP 4 family, predominantly catalyze conversion of AA to 20-hydroxyeicosatetraenoic acid (20-HETE) while the CYP epoxygenases generate mainly epoxyeicosatrienoic acids (EETs). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 142-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 17164129-4 2007 A number of these enzymes, including members of the CYP 4 family, predominantly catalyze conversion of AA to 20-hydroxyeicosatetraenoic acid (20-HETE) while the CYP epoxygenases generate mainly epoxyeicosatrienoic acids (EETs). epoxyeicosatrienoic acids 194-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 17164129-4 2007 A number of these enzymes, including members of the CYP 4 family, predominantly catalyze conversion of AA to 20-hydroxyeicosatetraenoic acid (20-HETE) while the CYP epoxygenases generate mainly epoxyeicosatrienoic acids (EETs). epoxyeicosatrienoic acids 194-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 17164129-4 2007 A number of these enzymes, including members of the CYP 4 family, predominantly catalyze conversion of AA to 20-hydroxyeicosatetraenoic acid (20-HETE) while the CYP epoxygenases generate mainly epoxyeicosatrienoic acids (EETs). eets 221-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 17164129-4 2007 A number of these enzymes, including members of the CYP 4 family, predominantly catalyze conversion of AA to 20-hydroxyeicosatetraenoic acid (20-HETE) while the CYP epoxygenases generate mainly epoxyeicosatrienoic acids (EETs). eets 221-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 17164129-5 2007 This review will focus on the emerging roles of inhibitors of eicosanoid production with emphasis on the CYP pathways, in the regulation of angiogenesis and tumor growth. Eicosanoids 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-108 17164132-5 2007 Current research suggests important differences with respect to the cardiovascular actions of specific CYP mediated arachidonic acid metabolites. Arachidonic Acid 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 17164132-6 2007 For example, CYP mediated hydroxylation of AA produces 20-hydroxyeicosatetraenoic acid (20-HETE) which has detrimental effects in the heart during ischemia, pro-inflammatory effects during reperfusion and potent vasoconstrictor effects in the coronary circulation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 55-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 17164132-6 2007 For example, CYP mediated hydroxylation of AA produces 20-hydroxyeicosatetraenoic acid (20-HETE) which has detrimental effects in the heart during ischemia, pro-inflammatory effects during reperfusion and potent vasoconstrictor effects in the coronary circulation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 88-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 18192961-8 2007 Loperamide is metabolized by the cytochrome P450 (CYP) system and is a substrate for the CYP3A4 isoenzyme. Loperamide 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 18192961-8 2007 Loperamide is metabolized by the cytochrome P450 (CYP) system and is a substrate for the CYP3A4 isoenzyme. Loperamide 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 17164132-7 2007 Conversely, epoxidation of AA by CYP enzymes generates 5,6-, 8,9-, 11,12- and 14,15-epoxyeicosatrienoic acids (EETs) that have been shown to reduce ischemia-reperfusion injury, have potent anti-inflammatory effects within the vasculature, and are potent vasodilators in the coronary circulation. 5,6-, 8,9-, 11,12- and 14,15-epoxyeicosatrienoic acids 55-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 17164132-7 2007 Conversely, epoxidation of AA by CYP enzymes generates 5,6-, 8,9-, 11,12- and 14,15-epoxyeicosatrienoic acids (EETs) that have been shown to reduce ischemia-reperfusion injury, have potent anti-inflammatory effects within the vasculature, and are potent vasodilators in the coronary circulation. 14,15-epoxy-5,8,11-eicosatrienoic acid 111-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 17164133-1 2007 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases the expression of which is determined by hemodynamic and pharmacological stimuli as well as by hypoxia. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 17164133-1 2007 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases the expression of which is determined by hemodynamic and pharmacological stimuli as well as by hypoxia. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 17164133-1 2007 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases the expression of which is determined by hemodynamic and pharmacological stimuli as well as by hypoxia. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 17164133-1 2007 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases the expression of which is determined by hemodynamic and pharmacological stimuli as well as by hypoxia. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 17164133-1 2007 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases the expression of which is determined by hemodynamic and pharmacological stimuli as well as by hypoxia. Arachidonic Acid 52-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 17164133-1 2007 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases the expression of which is determined by hemodynamic and pharmacological stimuli as well as by hypoxia. Arachidonic Acid 52-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 17202847-5 2006 Furthermore, SOCE is regulated by cytochrome P-450, as demonstrated by the fact that the products of cytochrome P-450 activity (14,15 EET) stimulated SOCE while econazole, an inhibitor of cytochrome P450, suppressed BK-stimulated Ca2+ entry. Econazole 161-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 17520800-3 2007 This review focuses on the flavonoid effects on cytochrome P450 (CYP) enzyme CYP1, 2E1, 3A4 and 19. Flavonoids 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 17520800-3 2007 This review focuses on the flavonoid effects on cytochrome P450 (CYP) enzyme CYP1, 2E1, 3A4 and 19. Flavonoids 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 17520800-4 2007 Flavonoids alter CYPs by various mechanisms, including the stimulation of gene expression via specific receptors and/or CYP protein, or mRNA stabilization and so on. Flavonoids 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 17202847-5 2006 Furthermore, SOCE is regulated by cytochrome P-450, as demonstrated by the fact that the products of cytochrome P-450 activity (14,15 EET) stimulated SOCE while econazole, an inhibitor of cytochrome P450, suppressed BK-stimulated Ca2+ entry. Econazole 161-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-117 17177535-0 2006 Metalloporphyrins as biomimetic models for cytochrome p-450 in the oxidation of atrazine. Metalloporphyrins 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 17177535-0 2006 Metalloporphyrins as biomimetic models for cytochrome p-450 in the oxidation of atrazine. Atrazine 80-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 17177535-1 2006 The aim of this work was to evaluate whether metalloporphyrin models could mimic the action of cytochrome P-450 in the oxidation of atrazine, a herbicide. Metalloporphyrins 45-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 17177535-1 2006 The aim of this work was to evaluate whether metalloporphyrin models could mimic the action of cytochrome P-450 in the oxidation of atrazine, a herbicide. Atrazine 132-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 17177535-4 2006 Yields as high as 32% were obtained for the Mn(TFPP)Cl/PhIO system, which shows that these catalysts can mimic both the in vivo and the in vitro action of cytochrome P-450, with formation of the metabolites DEA and DIA. dea 207-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 17177535-4 2006 Yields as high as 32% were obtained for the Mn(TFPP)Cl/PhIO system, which shows that these catalysts can mimic both the in vivo and the in vitro action of cytochrome P-450, with formation of the metabolites DEA and DIA. 4-(4-dihexadecylaminostyryl)-N-methylpyridium 215-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 17084997-0 2006 Ethanol oxidation into acetaldehyde by 16 recombinant human cytochrome P450 isoforms: role of CYP2C isoforms in human liver microsomes. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 17178892-6 2006 When MCF-7 cells were treated for 2 hours with the (+)-BP-7,8-diol, cigarette smoke increased dose dependently the formation of (-)-anti-BPDE-dG and decreased the cytochrome P450 (CYP)-dependent formation of (+)-r-7,t-8-dihydroxy-c-9,10-oxy-7,8,9,10-tetrahydro-BP the adduct. benzo(a)pyrene 7,8-dihydrodiol 51-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-178 17178892-6 2006 When MCF-7 cells were treated for 2 hours with the (+)-BP-7,8-diol, cigarette smoke increased dose dependently the formation of (-)-anti-BPDE-dG and decreased the cytochrome P450 (CYP)-dependent formation of (+)-r-7,t-8-dihydroxy-c-9,10-oxy-7,8,9,10-tetrahydro-BP the adduct. benzo(a)pyrene 7,8-dihydrodiol 51-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 17178892-6 2006 When MCF-7 cells were treated for 2 hours with the (+)-BP-7,8-diol, cigarette smoke increased dose dependently the formation of (-)-anti-BPDE-dG and decreased the cytochrome P450 (CYP)-dependent formation of (+)-r-7,t-8-dihydroxy-c-9,10-oxy-7,8,9,10-tetrahydro-BP the adduct. (-)-anti-bpde-dg 128-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 17178892-6 2006 When MCF-7 cells were treated for 2 hours with the (+)-BP-7,8-diol, cigarette smoke increased dose dependently the formation of (-)-anti-BPDE-dG and decreased the cytochrome P450 (CYP)-dependent formation of (+)-r-7,t-8-dihydroxy-c-9,10-oxy-7,8,9,10-tetrahydro-BP the adduct. (+)-r-7,t-8-dihydroxy-c-9,10-oxy-7,8,9,10-tetrahydro-bp 208-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-178 17178892-6 2006 When MCF-7 cells were treated for 2 hours with the (+)-BP-7,8-diol, cigarette smoke increased dose dependently the formation of (-)-anti-BPDE-dG and decreased the cytochrome P450 (CYP)-dependent formation of (+)-r-7,t-8-dihydroxy-c-9,10-oxy-7,8,9,10-tetrahydro-BP the adduct. (+)-r-7,t-8-dihydroxy-c-9,10-oxy-7,8,9,10-tetrahydro-bp 208-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 17084997-0 2006 Ethanol oxidation into acetaldehyde by 16 recombinant human cytochrome P450 isoforms: role of CYP2C isoforms in human liver microsomes. Acetaldehyde 23-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 17178266-1 2006 OBJECTIVES: Rosiglitazone is metabolically inactivated predominantly via the cytochrome P450 (CYP) enzyme CYP2C8. Rosiglitazone 12-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 16978758-0 2006 In vitro metabolism of tributyltin and triphenyltin by human cytochrome P-450 isoforms. tributyltin 23-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 16978758-0 2006 In vitro metabolism of tributyltin and triphenyltin by human cytochrome P-450 isoforms. triphenyltin 39-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 16978758-2 2006 We used human hepatic cytochrome P-450 (CYP) systems to confirm the specific CYP(s) involved in the in vitro metabolism of tributyltin and triphenyltin. tributyltin 123-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 16978758-2 2006 We used human hepatic cytochrome P-450 (CYP) systems to confirm the specific CYP(s) involved in the in vitro metabolism of tributyltin and triphenyltin. tributyltin 123-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 16978758-2 2006 We used human hepatic cytochrome P-450 (CYP) systems to confirm the specific CYP(s) involved in the in vitro metabolism of tributyltin and triphenyltin. tributyltin 123-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 16978758-2 2006 We used human hepatic cytochrome P-450 (CYP) systems to confirm the specific CYP(s) involved in the in vitro metabolism of tributyltin and triphenyltin. triphenyltin 139-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 16978758-2 2006 We used human hepatic cytochrome P-450 (CYP) systems to confirm the specific CYP(s) involved in the in vitro metabolism of tributyltin and triphenyltin. triphenyltin 139-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 16978758-2 2006 We used human hepatic cytochrome P-450 (CYP) systems to confirm the specific CYP(s) involved in the in vitro metabolism of tributyltin and triphenyltin. triphenyltin 139-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 17109460-1 2006 The ongoing interest in very efficient systems for the imitation of cytochrome P-450-dependent monooxygenase reactions, consisting of metalloporphyrin and oxygen donor, prompted us to develop a method to compare the catalytic activity of soluble metalloporphyrins with those which have been immobilised on different silica surfaces. Metalloporphyrins 134-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 17109460-1 2006 The ongoing interest in very efficient systems for the imitation of cytochrome P-450-dependent monooxygenase reactions, consisting of metalloporphyrin and oxygen donor, prompted us to develop a method to compare the catalytic activity of soluble metalloporphyrins with those which have been immobilised on different silica surfaces. Oxygen 99-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 17109460-1 2006 The ongoing interest in very efficient systems for the imitation of cytochrome P-450-dependent monooxygenase reactions, consisting of metalloporphyrin and oxygen donor, prompted us to develop a method to compare the catalytic activity of soluble metalloporphyrins with those which have been immobilised on different silica surfaces. Metalloporphyrins 246-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 17109460-1 2006 The ongoing interest in very efficient systems for the imitation of cytochrome P-450-dependent monooxygenase reactions, consisting of metalloporphyrin and oxygen donor, prompted us to develop a method to compare the catalytic activity of soluble metalloporphyrins with those which have been immobilised on different silica surfaces. Silicon Dioxide 316-322 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 17296539-7 2006 Rasagiline is almost completely eliminated by oxidative metabolism (catalyzed by cytochrome P-450 [CYP] isozyme 1A2) followed by renal excretion of conjugated parent compound and metabolites. rasagiline 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-115 17178266-1 2006 OBJECTIVES: Rosiglitazone is metabolically inactivated predominantly via the cytochrome P450 (CYP) enzyme CYP2C8. Rosiglitazone 12-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 17178267-0 2006 ABCB1 and cytochrome P450 genotypes and phenotypes: influence on methadone plasma levels and response to treatment. Methadone 65-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 17178267-1 2006 BACKGROUND AND OBJECTIVE: The in vivo implication of various cytochrome P450 (CYP) isoforms and of P-glycoprotein on methadone kinetics is unclear. Methadone 117-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 17178267-12 2006 CONCLUSION: In vivo, CYP3A4 and CYP2B6 are the major CYP isoforms involved in methadone metabolism, with CYP2D6 contributing to a minor extent. Methadone 78-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 16997912-0 2006 CYP4F enzymes are the major enzymes in human liver microsomes that catalyze the O-demethylation of the antiparasitic prodrug DB289 [2,5-bis(4-amidinophenyl)furan-bis-O-methylamidoxime]. pafuramidine 125-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-5 16997912-0 2006 CYP4F enzymes are the major enzymes in human liver microsomes that catalyze the O-demethylation of the antiparasitic prodrug DB289 [2,5-bis(4-amidinophenyl)furan-bis-O-methylamidoxime]. pafuramidine 132-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-5 17080490-1 2006 The use of CE for rapid assessment of metabolic stability of drugs with cytochrome P450 (CYP) enzymes, based on relative rates of reduced nicotinamide adenine dinucleotide phosphate (NADPH) consumption and nicotinamide adenine dinucleotide phosphate (NADP) production, was investigated. NADP 138-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 17080490-1 2006 The use of CE for rapid assessment of metabolic stability of drugs with cytochrome P450 (CYP) enzymes, based on relative rates of reduced nicotinamide adenine dinucleotide phosphate (NADPH) consumption and nicotinamide adenine dinucleotide phosphate (NADP) production, was investigated. NADP 138-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 17080490-1 2006 The use of CE for rapid assessment of metabolic stability of drugs with cytochrome P450 (CYP) enzymes, based on relative rates of reduced nicotinamide adenine dinucleotide phosphate (NADPH) consumption and nicotinamide adenine dinucleotide phosphate (NADP) production, was investigated. NADP 183-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 17080490-1 2006 The use of CE for rapid assessment of metabolic stability of drugs with cytochrome P450 (CYP) enzymes, based on relative rates of reduced nicotinamide adenine dinucleotide phosphate (NADPH) consumption and nicotinamide adenine dinucleotide phosphate (NADP) production, was investigated. NADP 183-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 17080490-1 2006 The use of CE for rapid assessment of metabolic stability of drugs with cytochrome P450 (CYP) enzymes, based on relative rates of reduced nicotinamide adenine dinucleotide phosphate (NADPH) consumption and nicotinamide adenine dinucleotide phosphate (NADP) production, was investigated. NADP 206-249 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 17080490-1 2006 The use of CE for rapid assessment of metabolic stability of drugs with cytochrome P450 (CYP) enzymes, based on relative rates of reduced nicotinamide adenine dinucleotide phosphate (NADPH) consumption and nicotinamide adenine dinucleotide phosphate (NADP) production, was investigated. NADP 206-249 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 17080490-1 2006 The use of CE for rapid assessment of metabolic stability of drugs with cytochrome P450 (CYP) enzymes, based on relative rates of reduced nicotinamide adenine dinucleotide phosphate (NADPH) consumption and nicotinamide adenine dinucleotide phosphate (NADP) production, was investigated. NADP 183-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 17080490-1 2006 The use of CE for rapid assessment of metabolic stability of drugs with cytochrome P450 (CYP) enzymes, based on relative rates of reduced nicotinamide adenine dinucleotide phosphate (NADPH) consumption and nicotinamide adenine dinucleotide phosphate (NADP) production, was investigated. NADP 183-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 17190370-1 2006 OBJECTIVE: Lansoprazole and tacrolimus are substrates of ATP binding cassette (ABC) transporters such as P-glycoprotein (ABCBI/multidrug resistance 1) and cytochrome P450 (CYP). Lansoprazole 11-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 17190370-1 2006 OBJECTIVE: Lansoprazole and tacrolimus are substrates of ATP binding cassette (ABC) transporters such as P-glycoprotein (ABCBI/multidrug resistance 1) and cytochrome P450 (CYP). Lansoprazole 11-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 17190370-1 2006 OBJECTIVE: Lansoprazole and tacrolimus are substrates of ATP binding cassette (ABC) transporters such as P-glycoprotein (ABCBI/multidrug resistance 1) and cytochrome P450 (CYP). Tacrolimus 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 17190370-1 2006 OBJECTIVE: Lansoprazole and tacrolimus are substrates of ATP binding cassette (ABC) transporters such as P-glycoprotein (ABCBI/multidrug resistance 1) and cytochrome P450 (CYP). Tacrolimus 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 16835590-8 2006 CYP4F3 and CYP4F2 catalyse the inactivation of leukotriene B4 (LTB4), a potent mediator of inflammation responsible for recruitment and activation of neutrophils. Leukotriene B4 47-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 16980726-0 2006 Hydroxyeicosatetraenoic acids released through the cytochrome P-450 pathway regulate 3T6 fibroblast growth. Hydroxyeicosatetraenoic Acids 0-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 16980726-4 2006 Our results show that 3T6 fibroblasts synthesized hydroxyeicosatetraenoic acids (HETEs) such as 12-HETE through the cytochrome P-450 (CYP450) pathway. Hydroxyeicosatetraenoic Acids 50-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 16980726-4 2006 Our results show that 3T6 fibroblasts synthesized hydroxyeicosatetraenoic acids (HETEs) such as 12-HETE through the cytochrome P-450 (CYP450) pathway. Hydroxyeicosatetraenoic Acids 50-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-140 16980726-4 2006 Our results show that 3T6 fibroblasts synthesized hydroxyeicosatetraenoic acids (HETEs) such as 12-HETE through the cytochrome P-450 (CYP450) pathway. Hydroxyeicosatetraenoic Acids 81-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 16980726-4 2006 Our results show that 3T6 fibroblasts synthesized hydroxyeicosatetraenoic acids (HETEs) such as 12-HETE through the cytochrome P-450 (CYP450) pathway. Hydroxyeicosatetraenoic Acids 81-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-140 16980726-4 2006 Our results show that 3T6 fibroblasts synthesized hydroxyeicosatetraenoic acids (HETEs) such as 12-HETE through the cytochrome P-450 (CYP450) pathway. 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid 96-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 16980726-4 2006 Our results show that 3T6 fibroblasts synthesized hydroxyeicosatetraenoic acids (HETEs) such as 12-HETE through the cytochrome P-450 (CYP450) pathway. 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid 96-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-140 16980726-6 2006 In contrast, we observed that CYP450 inhibitors such as SKF-525A, 17-octadecynoic acid, 1-aminobenzotriazole, and 6-(2-propargyloxyphenyl)hexanoic acid reduced 12(S)-HETE levels, 3T6 fibroblast growth, and DNA synthesis induced by FBS. Proadifen 56-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-36 16980726-6 2006 In contrast, we observed that CYP450 inhibitors such as SKF-525A, 17-octadecynoic acid, 1-aminobenzotriazole, and 6-(2-propargyloxyphenyl)hexanoic acid reduced 12(S)-HETE levels, 3T6 fibroblast growth, and DNA synthesis induced by FBS. 17-octadecynoic acid 66-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-36 16980726-6 2006 In contrast, we observed that CYP450 inhibitors such as SKF-525A, 17-octadecynoic acid, 1-aminobenzotriazole, and 6-(2-propargyloxyphenyl)hexanoic acid reduced 12(S)-HETE levels, 3T6 fibroblast growth, and DNA synthesis induced by FBS. 1-aminobenzotriazole 88-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-36 16980726-6 2006 In contrast, we observed that CYP450 inhibitors such as SKF-525A, 17-octadecynoic acid, 1-aminobenzotriazole, and 6-(2-propargyloxyphenyl)hexanoic acid reduced 12(S)-HETE levels, 3T6 fibroblast growth, and DNA synthesis induced by FBS. 6-(2-propargyloxyphenyl)hexanoic acid 114-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-36 17159771-2 2006 Recently, we found that after cytochrome P450 (CYP)-mediated oxidation ellipticine forms covalent DNA adducts. ellipticine 71-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 17159771-2 2006 Recently, we found that after cytochrome P450 (CYP)-mediated oxidation ellipticine forms covalent DNA adducts. ellipticine 71-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 17159771-3 2006 Ellipticine oxidation by isolated CYP and its binding to DNA is the target of this study. ellipticine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 17159771-6 2006 RESULTS: Purified CYP enzymes reconstituted with NADPH:CYP reductase oxidized ellipticine to up to five metabolites, 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine and ellipticine N(2)-oxide. NADP 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 17159771-6 2006 RESULTS: Purified CYP enzymes reconstituted with NADPH:CYP reductase oxidized ellipticine to up to five metabolites, 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine and ellipticine N(2)-oxide. NADP 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 17159771-6 2006 RESULTS: Purified CYP enzymes reconstituted with NADPH:CYP reductase oxidized ellipticine to up to five metabolites, 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine and ellipticine N(2)-oxide. ellipticine 78-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 17159771-6 2006 RESULTS: Purified CYP enzymes reconstituted with NADPH:CYP reductase oxidized ellipticine to up to five metabolites, 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine and ellipticine N(2)-oxide. ellipticine 78-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 17159771-6 2006 RESULTS: Purified CYP enzymes reconstituted with NADPH:CYP reductase oxidized ellipticine to up to five metabolites, 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine and ellipticine N(2)-oxide. 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine 117-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 17159771-6 2006 RESULTS: Purified CYP enzymes reconstituted with NADPH:CYP reductase oxidized ellipticine to up to five metabolites, 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine and ellipticine N(2)-oxide. 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine 117-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 17159771-6 2006 RESULTS: Purified CYP enzymes reconstituted with NADPH:CYP reductase oxidized ellipticine to up to five metabolites, 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine and ellipticine N(2)-oxide. ellipticine n(2)-oxide 180-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 17159771-6 2006 RESULTS: Purified CYP enzymes reconstituted with NADPH:CYP reductase oxidized ellipticine to up to five metabolites, 7-hydroxy-, 9-hydroxy-, 12-hydroxy-, 13-hydroxyellipticine and ellipticine N(2)-oxide. ellipticine n(2)-oxide 180-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 17159771-10 2006 CONCLUSIONS: The results showed that the system of purified CYPs reconstituted with NADPH: CYP reductase proved for ellipticine oxidation provide a true reflection of the situation in the microsomal membrane. NADP 84-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 17159771-10 2006 CONCLUSIONS: The results showed that the system of purified CYPs reconstituted with NADPH: CYP reductase proved for ellipticine oxidation provide a true reflection of the situation in the microsomal membrane. ellipticine 116-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 17164372-0 2006 Effects of repeated green tea catechin administration on human cytochrome P450 activity. Catechin 30-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 17164372-2 2006 We conducted this clinical study to determine the effect of repeated green tea catechin administration on human cytochrome P450 (CYP) enzyme activities. Catechin 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-127 17164372-2 2006 We conducted this clinical study to determine the effect of repeated green tea catechin administration on human cytochrome P450 (CYP) enzyme activities. Catechin 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-132 16926051-0 2006 Cytochrome P450 4F subfamily: at the crossroads of eicosanoid and drug metabolism. Eicosanoids 51-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-18 16926051-1 2006 The cytochrome P450 4F (CYP4F) subfamily has over the last few years come to be recognized for its dual role in modulating the concentrations of eicosanoids during inflammation as well as in the metabolism of clinically significant drugs. Eicosanoids 145-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-22 16926051-1 2006 The cytochrome P450 4F (CYP4F) subfamily has over the last few years come to be recognized for its dual role in modulating the concentrations of eicosanoids during inflammation as well as in the metabolism of clinically significant drugs. Eicosanoids 145-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-29 16926051-2 2006 The first CYP4F was identified because it catalyzed the hydroxylation of leukotriene B(4) (LTB(4)) and since then many additional members of this subfamily have been documented for their distinct catalytic roles and functional significance. Leukotriene B4 73-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-15 16926051-5 2006 From this standpoint variations in relative CYP4F levels in humans may have direct influence on the metabolic outcome through their ability to generate and/or degrade bioactive eicosanoids or therapeutic agents. Eicosanoids 177-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-49 16926051-6 2006 This review covers the enzymatic characteristics and regulatory properties of human and rodent CYP4F isoforms and their physiological relevance to major pathways in eicosanoid and drug metabolism. Eicosanoids 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-100 16945988-1 2006 The present study investigated the role of specific human cytochrome P450 (CYP) enzymes in the in vitro metabolism of valproic acid (VPA) by a complementary approach that used individual cDNA-expressed CYP enzymes, chemical inhibitors of specific CYP enzymes, CYP-specific inhibitory monoclonal antibodies (MAbs), individual human hepatic microsomes, and correlational analysis. Valproic Acid 118-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 16945988-1 2006 The present study investigated the role of specific human cytochrome P450 (CYP) enzymes in the in vitro metabolism of valproic acid (VPA) by a complementary approach that used individual cDNA-expressed CYP enzymes, chemical inhibitors of specific CYP enzymes, CYP-specific inhibitory monoclonal antibodies (MAbs), individual human hepatic microsomes, and correlational analysis. Valproic Acid 118-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 16945988-1 2006 The present study investigated the role of specific human cytochrome P450 (CYP) enzymes in the in vitro metabolism of valproic acid (VPA) by a complementary approach that used individual cDNA-expressed CYP enzymes, chemical inhibitors of specific CYP enzymes, CYP-specific inhibitory monoclonal antibodies (MAbs), individual human hepatic microsomes, and correlational analysis. Valproic Acid 133-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 16945988-1 2006 The present study investigated the role of specific human cytochrome P450 (CYP) enzymes in the in vitro metabolism of valproic acid (VPA) by a complementary approach that used individual cDNA-expressed CYP enzymes, chemical inhibitors of specific CYP enzymes, CYP-specific inhibitory monoclonal antibodies (MAbs), individual human hepatic microsomes, and correlational analysis. Valproic Acid 133-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 16835590-8 2006 CYP4F3 and CYP4F2 catalyse the inactivation of leukotriene B4 (LTB4), a potent mediator of inflammation responsible for recruitment and activation of neutrophils. Leukotriene B4 63-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 16940989-1 2006 BACKGROUND AND PURPOSE: The types of hepatic microsomal cytochrome P450 (CYP) isozymes responsible for the metabolism of metformin in humans and rats have not been published to date. Metformin 121-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 16778795-2 2006 Metabolic transformation of all-trans RA to 4-hydroxylated RA appears to be primarily catalyzed by the cytochrome P450 (CYP) 26AI. Tretinoin 38-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 16778795-2 2006 Metabolic transformation of all-trans RA to 4-hydroxylated RA appears to be primarily catalyzed by the cytochrome P450 (CYP) 26AI. Tretinoin 38-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 16778795-2 2006 Metabolic transformation of all-trans RA to 4-hydroxylated RA appears to be primarily catalyzed by the cytochrome P450 (CYP) 26AI. Tretinoin 59-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 16778795-2 2006 Metabolic transformation of all-trans RA to 4-hydroxylated RA appears to be primarily catalyzed by the cytochrome P450 (CYP) 26AI. Tretinoin 59-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 16965820-0 2006 New approach to the activation of anti-cancer pro-drugs by metalloporphyrin-based cytochrome P450 mimics in all-aqueous biologically relevant system. Metalloporphyrins 59-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 16824612-4 2006 Furthermore, mounting evidence points to the role of endogenous CYP metabolites, such as epoxyeicosatrienoic acids (EETs), hydroxyeicosatetraenoic acids (HETEs), prostacyclin (PGI(2)), aldosterone, and sex hormones, in the maintenance of cardiovascular health. Aldosterone 185-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 16943206-0 2006 Cytochrome P450 active site plasticity: attenuation of imidazole binding in cytochrome P450(cam) by an L244A mutation. imidazole 55-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 16943206-0 2006 Cytochrome P450 active site plasticity: attenuation of imidazole binding in cytochrome P450(cam) by an L244A mutation. imidazole 55-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 16857327-0 2006 Characterization of the cytochrome P450 enzymes involved in the metabolism of a new cardioprotective agent KR-33028. Krypton 107-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 16857327-2 2006 This study was performed to characterize the cytochrome P450 (CYP) enzymes that are involved in the metabolism of KR-33028. Krypton 114-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 16857327-2 2006 This study was performed to characterize the cytochrome P450 (CYP) enzymes that are involved in the metabolism of KR-33028. Krypton 114-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 16857327-4 2006 Among the nine c-DNA expressed CYP isoforms tested, KR-33028 was 5-hydroxylated by CYP3A4 and 7-hydroxylated by CYP1A2, CYP3A4, and CYP2C19. Krypton 52-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 16857327-7 2006 Correlation analysis between the known CYP enzyme activities and the rates of the formation of 5-hydroxy- and 7-hydroxy-KR-33028 in the 16 human liver microsomes has showed significant correlations with CYP3A4-mediated midazolam 1"-hydroxylation and CYP1A2-mediated phenacetin O-deethylation, respectively. 5-hydroxy- and 7-hydroxy-kr 95-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 16857327-7 2006 Correlation analysis between the known CYP enzyme activities and the rates of the formation of 5-hydroxy- and 7-hydroxy-KR-33028 in the 16 human liver microsomes has showed significant correlations with CYP3A4-mediated midazolam 1"-hydroxylation and CYP1A2-mediated phenacetin O-deethylation, respectively. Midazolam 219-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 16857327-7 2006 Correlation analysis between the known CYP enzyme activities and the rates of the formation of 5-hydroxy- and 7-hydroxy-KR-33028 in the 16 human liver microsomes has showed significant correlations with CYP3A4-mediated midazolam 1"-hydroxylation and CYP1A2-mediated phenacetin O-deethylation, respectively. Phenacetin 266-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 16857327-9 2006 The kinetics of the major biotransformation of KR-33028 were studied: CYP3A4 mediated the formation of 5-hydroxy-KR-33028 from KR-33028 with Cl(int)=0.22microl/min/pmol CYP. 5-hydroxy-kr 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 16857327-9 2006 The kinetics of the major biotransformation of KR-33028 were studied: CYP3A4 mediated the formation of 5-hydroxy-KR-33028 from KR-33028 with Cl(int)=0.22microl/min/pmol CYP. Krypton 47-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 16857327-10 2006 The intrinsic clearance for 7-hydroxy-KR-33028 formation by CYP1A2, CYP2C19, and CYP3A4 were 0.26, 0.19, and 0.03microl/min/pmol CYP, respectively. 7-hydroxy-kr 28-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 16772608-2 2006 To determine whether frequent functional variants of genes coding for candidate cytochrome P450 (CYP) isoenzymes involved in clopidogrel metabolic activation (CYP2C19*2, CYP2B6*5, CYP1A2*1F, and CYP3A5*3 variants) influence the platelet responsiveness to clopidogrel, we conducted a prospective pharmacogenetic study in 28 healthy white male volunteers treated for 7 days with clopidogrel 75 mg/d. Clopidogrel 125-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 16772608-2 2006 To determine whether frequent functional variants of genes coding for candidate cytochrome P450 (CYP) isoenzymes involved in clopidogrel metabolic activation (CYP2C19*2, CYP2B6*5, CYP1A2*1F, and CYP3A5*3 variants) influence the platelet responsiveness to clopidogrel, we conducted a prospective pharmacogenetic study in 28 healthy white male volunteers treated for 7 days with clopidogrel 75 mg/d. Clopidogrel 125-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 16772608-2 2006 To determine whether frequent functional variants of genes coding for candidate cytochrome P450 (CYP) isoenzymes involved in clopidogrel metabolic activation (CYP2C19*2, CYP2B6*5, CYP1A2*1F, and CYP3A5*3 variants) influence the platelet responsiveness to clopidogrel, we conducted a prospective pharmacogenetic study in 28 healthy white male volunteers treated for 7 days with clopidogrel 75 mg/d. Clopidogrel 255-266 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 16772608-2 2006 To determine whether frequent functional variants of genes coding for candidate cytochrome P450 (CYP) isoenzymes involved in clopidogrel metabolic activation (CYP2C19*2, CYP2B6*5, CYP1A2*1F, and CYP3A5*3 variants) influence the platelet responsiveness to clopidogrel, we conducted a prospective pharmacogenetic study in 28 healthy white male volunteers treated for 7 days with clopidogrel 75 mg/d. Clopidogrel 255-266 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 16772608-2 2006 To determine whether frequent functional variants of genes coding for candidate cytochrome P450 (CYP) isoenzymes involved in clopidogrel metabolic activation (CYP2C19*2, CYP2B6*5, CYP1A2*1F, and CYP3A5*3 variants) influence the platelet responsiveness to clopidogrel, we conducted a prospective pharmacogenetic study in 28 healthy white male volunteers treated for 7 days with clopidogrel 75 mg/d. Clopidogrel 255-266 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 16772608-2 2006 To determine whether frequent functional variants of genes coding for candidate cytochrome P450 (CYP) isoenzymes involved in clopidogrel metabolic activation (CYP2C19*2, CYP2B6*5, CYP1A2*1F, and CYP3A5*3 variants) influence the platelet responsiveness to clopidogrel, we conducted a prospective pharmacogenetic study in 28 healthy white male volunteers treated for 7 days with clopidogrel 75 mg/d. Clopidogrel 255-266 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 16940989-1 2006 BACKGROUND AND PURPOSE: The types of hepatic microsomal cytochrome P450 (CYP) isozymes responsible for the metabolism of metformin in humans and rats have not been published to date. Metformin 121-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 16927149-0 2006 Monitoring cytochrome P-450 activity during rabeprazole treatment in patients with gastresophageal reflux disease. Rabeprazole 44-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 17018623-8 2006 Inhibition of SULTs and cytochrome P450 (CYP) enzymes by natural flavonoids blocked the antiproliferative activity of AF and the formation of AF-DNA adducts. Flavonoids 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 17018623-8 2006 Inhibition of SULTs and cytochrome P450 (CYP) enzymes by natural flavonoids blocked the antiproliferative activity of AF and the formation of AF-DNA adducts. Flavonoids 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 17018623-10 2006 Metabolism studies showed that AF can be oxidized by CYP at two amino groups to form N-hydroxyl metabolites that are substrates for bioactivation by SULTs. n-hydroxyl 85-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 138-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 138-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 162-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 162-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). N-desmethylvenlafaxine 211-233 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). N-desmethylvenlafaxine 211-233 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). N-desmethylvenlafaxine 235-238 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). N-desmethylvenlafaxine 235-238 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. N-acetylserotonin 84-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 17197724-1 2006 Ellipticine is an antineoplastic agent, whose mode of action is based mainly on DNA intercalation, inhibition of topoisomerase II and formation of DNA adducts mediated by cytochrome P450 (CYP). ellipticine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-186 17197724-1 2006 Ellipticine is an antineoplastic agent, whose mode of action is based mainly on DNA intercalation, inhibition of topoisomerase II and formation of DNA adducts mediated by cytochrome P450 (CYP). ellipticine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-191 17225875-5 2006 CYP enzymes, including CYP2D6, participate in the metabolism of some porphyrinogenic drugs, leading to the deregulation of heme biosynthesis. Heme 123-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 16914311-6 2006 These versions suggest that some cytochrome P-450"s catalyze the introduction of both oxygen atoms of dioxygen into an appropriate sterol precursor. Oxygen 86-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 16914311-6 2006 These versions suggest that some cytochrome P-450"s catalyze the introduction of both oxygen atoms of dioxygen into an appropriate sterol precursor. Oxygen 102-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 16914311-6 2006 These versions suggest that some cytochrome P-450"s catalyze the introduction of both oxygen atoms of dioxygen into an appropriate sterol precursor. Sterols 131-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. Melatonin 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. Melatonin 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. 6-sulfatoxymelatonin 31-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. N-acetylserotonin 84-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. 6-sulfatoxymelatonin 31-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 16380164-1 2006 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. amt6s 54-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. amt6s 54-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. N-acetylserotonin 65-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 17003850-1 2006 The metabolism of melatonin to 6-sulphatoxymelatonin (aMT6S) and N-acetylserotonin (NAS) is catalyzed by cytochrome-P450 (CYP) isozymes CYP1A2 and CYP2C19 respectively. N-acetylserotonin 65-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 16380164-1 2006 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 16380164-1 2006 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 16380164-1 2006 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 16380164-1 2006 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases. Arachidonic Acid 52-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 16380164-1 2006 Epoxyeicosatrienoic acids (EETs) are generated from arachidonic acid by cytochrome P450 (CYP) epoxygenases. Arachidonic Acid 52-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 16380164-3 2006 The activation of CYP epoxygenases in endothelial cells is an important step in the nitric oxide and prostacyclin (PGI2)-independent vasodilatation of several vascular beds and EETs have been identified as endothelium-derived hyperpolarizing factors (EDHFs). Nitric Oxide 84-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 16380164-3 2006 The activation of CYP epoxygenases in endothelial cells is an important step in the nitric oxide and prostacyclin (PGI2)-independent vasodilatation of several vascular beds and EETs have been identified as endothelium-derived hyperpolarizing factors (EDHFs). Epoprostenol 101-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 16380164-3 2006 The activation of CYP epoxygenases in endothelial cells is an important step in the nitric oxide and prostacyclin (PGI2)-independent vasodilatation of several vascular beds and EETs have been identified as endothelium-derived hyperpolarizing factors (EDHFs). Epoprostenol 115-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 16380164-3 2006 The activation of CYP epoxygenases in endothelial cells is an important step in the nitric oxide and prostacyclin (PGI2)-independent vasodilatation of several vascular beds and EETs have been identified as endothelium-derived hyperpolarizing factors (EDHFs). Biotin 242-249 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 16380164-3 2006 The activation of CYP epoxygenases in endothelial cells is an important step in the nitric oxide and prostacyclin (PGI2)-independent vasodilatation of several vascular beds and EETs have been identified as endothelium-derived hyperpolarizing factors (EDHFs). edhfs 251-256 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 16925400-3 2006 Upon binding of a substrate molecule (e.g., camphor), the absorption band of cytochrome P450 shifts to shorter wavelength. Camphor 44-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 16925400-4 2006 The event of camphor binding to a nanoparticle surface modified with cytochrome P450 protein receptors is monitored using UV-vis spectroscopy. Camphor 13-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 16802166-0 2006 Effect of psychotropic medication on the in vitro metabolism of buprenorphine in human cDNA-expressed cytochrome P450 enzymes. Buprenorphine 64-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-117 16867264-0 2006 Influences of 3-methylcholanthrene, phenobarbital and dexamethasone on xenobiotic metabolizing-related cytochrome P450 enzymes and steroidogenesis in human fetal adrenal cortical cells. Methylcholanthrene 14-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 16867264-0 2006 Influences of 3-methylcholanthrene, phenobarbital and dexamethasone on xenobiotic metabolizing-related cytochrome P450 enzymes and steroidogenesis in human fetal adrenal cortical cells. Phenobarbital 36-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 16867264-0 2006 Influences of 3-methylcholanthrene, phenobarbital and dexamethasone on xenobiotic metabolizing-related cytochrome P450 enzymes and steroidogenesis in human fetal adrenal cortical cells. Dexamethasone 54-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 16867264-11 2006 CONCLUSION: 3-Methylcholanthrene, phenobarbital or dexamethasone could interfere with the synthesis of cortisol, aldosterone and progesterone in primary human fetal adrenal cortical cells, which likely act through xenobiotic metabolizing-related cytochrome P450 isoform activation. Methylcholanthrene 12-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-261 16867264-11 2006 CONCLUSION: 3-Methylcholanthrene, phenobarbital or dexamethasone could interfere with the synthesis of cortisol, aldosterone and progesterone in primary human fetal adrenal cortical cells, which likely act through xenobiotic metabolizing-related cytochrome P450 isoform activation. Phenobarbital 34-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-261 16867264-11 2006 CONCLUSION: 3-Methylcholanthrene, phenobarbital or dexamethasone could interfere with the synthesis of cortisol, aldosterone and progesterone in primary human fetal adrenal cortical cells, which likely act through xenobiotic metabolizing-related cytochrome P450 isoform activation. Dexamethasone 51-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-261 16867264-11 2006 CONCLUSION: 3-Methylcholanthrene, phenobarbital or dexamethasone could interfere with the synthesis of cortisol, aldosterone and progesterone in primary human fetal adrenal cortical cells, which likely act through xenobiotic metabolizing-related cytochrome P450 isoform activation. Hydrocortisone 103-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-261 16867264-11 2006 CONCLUSION: 3-Methylcholanthrene, phenobarbital or dexamethasone could interfere with the synthesis of cortisol, aldosterone and progesterone in primary human fetal adrenal cortical cells, which likely act through xenobiotic metabolizing-related cytochrome P450 isoform activation. Progesterone 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-261 16890580-1 2006 OBJECTIVE: Our objective was to explore the relationships between imatinib pharmacokinetics and 9 allelic variants in 7 genes coding for adenosine triphosphate-binding cassette transporters (ABCB1 and ABCG2) and enzymes (cytochrome P450 [CYP] 2C9, CYP2C19, CYP2D6, CYP3A4, and CYP3A5) of putative relevance for imatinib. Imatinib Mesylate 66-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-246 16802166-1 2006 OBJECTIVE: The aim of the present study was to estimate the drug interaction potential of psychtropic medication on buprenorphine (BUP) N-dealkylation using cDNA-expressed cytochrome P450 (CYP) enzymes. Buprenorphine 116-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-187 16802166-1 2006 OBJECTIVE: The aim of the present study was to estimate the drug interaction potential of psychtropic medication on buprenorphine (BUP) N-dealkylation using cDNA-expressed cytochrome P450 (CYP) enzymes. Buprenorphine 116-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-192 16820944-15 2006 Inhibition of proinflammatory cytokines by curcumin may provide a novel approach to modulate the hepatic CYP function in sepsis. Curcumin 43-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-108 16937919-17 2006 Bz also inhibits the metabolism of several xenobiotics biotransformed by the cytochrome P450 system and its reactive metabolites react with fetal components in vivo. benzonidazole 0-2 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 16494911-7 2006 The present data indicating alterations in the expression of xenobiotic metabolizing CYPs during development following prenatal exposure to deltamethrin may be of significance as these CYP enzymes are not only involved in the neurobehavioral toxicity of deltamethrin but have a role in regulating the levels of ligands that modulate growth, differentiation, and neuroendocrine functions. decamethrin 140-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 16494911-7 2006 The present data indicating alterations in the expression of xenobiotic metabolizing CYPs during development following prenatal exposure to deltamethrin may be of significance as these CYP enzymes are not only involved in the neurobehavioral toxicity of deltamethrin but have a role in regulating the levels of ligands that modulate growth, differentiation, and neuroendocrine functions. decamethrin 254-266 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 16639745-1 2006 To examine the molecular basis of activity and regioselectivity of the clinically important human microsomal cytochrome P450 (CYP) monooxygenase 2C9 toward its substrate warfarin, 22 molecular dynamics simulations (3-5 ns each) were performed in the presence and absence of warfarin. Warfarin 170-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 16764555-2 2006 Three major enzymatic pathways, COX (cyclo-oxygenase), CYP450 (cytochrome P450) and LOX (lipoxygenase), are responsible for the metabolism of arachidonic acid metabolites to bioactive eicosanoids. Arachidonic Acid 142-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-61 16764555-2 2006 Three major enzymatic pathways, COX (cyclo-oxygenase), CYP450 (cytochrome P450) and LOX (lipoxygenase), are responsible for the metabolism of arachidonic acid metabolites to bioactive eicosanoids. Arachidonic Acid 142-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 16764555-2 2006 Three major enzymatic pathways, COX (cyclo-oxygenase), CYP450 (cytochrome P450) and LOX (lipoxygenase), are responsible for the metabolism of arachidonic acid metabolites to bioactive eicosanoids. Eicosanoids 184-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-61 16764555-2 2006 Three major enzymatic pathways, COX (cyclo-oxygenase), CYP450 (cytochrome P450) and LOX (lipoxygenase), are responsible for the metabolism of arachidonic acid metabolites to bioactive eicosanoids. Eicosanoids 184-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 16782090-0 2006 Identification of the cytochrome P450 enzymes responsible for the omega-hydroxylation of phytanic acid. Phytanic Acid 89-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 16782090-3 2006 With the use of specific inhibitors we now show that members of the cytochrome P450 (CYP450) family 4 class are responsible for phytanic acid omega-hydroxylation. Phytanic Acid 128-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 16782090-3 2006 With the use of specific inhibitors we now show that members of the cytochrome P450 (CYP450) family 4 class are responsible for phytanic acid omega-hydroxylation. Phytanic Acid 128-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-91 16782090-4 2006 Incubations with microsomes containing human recombinant CYP450s (Supersomes) revealed that multiple CYP450 enzymes of the family 4 class are able to omega-hydroxylate phytanic acid with the following order of efficiency: CYP4F3A>CYP4F3B>CYP4F2>CYP4A11. Phytanic Acid 168-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-63 16782090-4 2006 Incubations with microsomes containing human recombinant CYP450s (Supersomes) revealed that multiple CYP450 enzymes of the family 4 class are able to omega-hydroxylate phytanic acid with the following order of efficiency: CYP4F3A>CYP4F3B>CYP4F2>CYP4A11. Phytanic Acid 168-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 16886668-1 2006 Like most pharmaceutical agents, vitamin D analogs are subject to hepatic metabolism by a variety of cytochrome P450 (CYP)-based systems. Vitamin D 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 16886668-1 2006 Like most pharmaceutical agents, vitamin D analogs are subject to hepatic metabolism by a variety of cytochrome P450 (CYP)-based systems. Vitamin D 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 16639745-1 2006 To examine the molecular basis of activity and regioselectivity of the clinically important human microsomal cytochrome P450 (CYP) monooxygenase 2C9 toward its substrate warfarin, 22 molecular dynamics simulations (3-5 ns each) were performed in the presence and absence of warfarin. Warfarin 170-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 16639745-1 2006 To examine the molecular basis of activity and regioselectivity of the clinically important human microsomal cytochrome P450 (CYP) monooxygenase 2C9 toward its substrate warfarin, 22 molecular dynamics simulations (3-5 ns each) were performed in the presence and absence of warfarin. Warfarin 274-282 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 16639745-1 2006 To examine the molecular basis of activity and regioselectivity of the clinically important human microsomal cytochrome P450 (CYP) monooxygenase 2C9 toward its substrate warfarin, 22 molecular dynamics simulations (3-5 ns each) were performed in the presence and absence of warfarin. Warfarin 274-282 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 16641218-1 2006 Animal studies have shown that induction of cytochrome P450 (CYP) in the lung by oxygen exposure may result in the release of free radical oxidants and arachidonic acid metabolites, which can cause lung injury that is reduced by treatment with cimetidine, a CYP inhibitor. Oxygen 81-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 16864505-0 2006 Metabolism of cytostatically active 6-aminobenzo[c]phenanthridines by human and porcine hepatic microsomes and recombinant cytochrome P450 enzymes. 6-aminobenzo[c]phenanthridines 36-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-138 16713055-6 2006 These observations suggest that NOS2 can behave similarly to cytochrome P-450 in the catalysis of acetaldehyde formation from ethanol via the generation of alpha-hydroxyethyl radical when L-arginine is present. Acetaldehyde 98-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 16713055-6 2006 These observations suggest that NOS2 can behave similarly to cytochrome P-450 in the catalysis of acetaldehyde formation from ethanol via the generation of alpha-hydroxyethyl radical when L-arginine is present. Ethanol 126-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 16713055-6 2006 These observations suggest that NOS2 can behave similarly to cytochrome P-450 in the catalysis of acetaldehyde formation from ethanol via the generation of alpha-hydroxyethyl radical when L-arginine is present. alpha-hydroxyethyl radical 156-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 16713055-6 2006 These observations suggest that NOS2 can behave similarly to cytochrome P-450 in the catalysis of acetaldehyde formation from ethanol via the generation of alpha-hydroxyethyl radical when L-arginine is present. Arginine 188-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 16641218-1 2006 Animal studies have shown that induction of cytochrome P450 (CYP) in the lung by oxygen exposure may result in the release of free radical oxidants and arachidonic acid metabolites, which can cause lung injury that is reduced by treatment with cimetidine, a CYP inhibitor. Oxygen 81-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 16641218-1 2006 Animal studies have shown that induction of cytochrome P450 (CYP) in the lung by oxygen exposure may result in the release of free radical oxidants and arachidonic acid metabolites, which can cause lung injury that is reduced by treatment with cimetidine, a CYP inhibitor. Oxygen 81-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-261 16641218-1 2006 Animal studies have shown that induction of cytochrome P450 (CYP) in the lung by oxygen exposure may result in the release of free radical oxidants and arachidonic acid metabolites, which can cause lung injury that is reduced by treatment with cimetidine, a CYP inhibitor. Arachidonic Acid 152-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 16641218-1 2006 Animal studies have shown that induction of cytochrome P450 (CYP) in the lung by oxygen exposure may result in the release of free radical oxidants and arachidonic acid metabolites, which can cause lung injury that is reduced by treatment with cimetidine, a CYP inhibitor. Arachidonic Acid 152-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 16641218-1 2006 Animal studies have shown that induction of cytochrome P450 (CYP) in the lung by oxygen exposure may result in the release of free radical oxidants and arachidonic acid metabolites, which can cause lung injury that is reduced by treatment with cimetidine, a CYP inhibitor. Arachidonic Acid 152-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-261 16641218-1 2006 Animal studies have shown that induction of cytochrome P450 (CYP) in the lung by oxygen exposure may result in the release of free radical oxidants and arachidonic acid metabolites, which can cause lung injury that is reduced by treatment with cimetidine, a CYP inhibitor. Cimetidine 244-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 16641218-1 2006 Animal studies have shown that induction of cytochrome P450 (CYP) in the lung by oxygen exposure may result in the release of free radical oxidants and arachidonic acid metabolites, which can cause lung injury that is reduced by treatment with cimetidine, a CYP inhibitor. Cimetidine 244-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 16641218-1 2006 Animal studies have shown that induction of cytochrome P450 (CYP) in the lung by oxygen exposure may result in the release of free radical oxidants and arachidonic acid metabolites, which can cause lung injury that is reduced by treatment with cimetidine, a CYP inhibitor. Cimetidine 244-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-261 16510126-0 2006 Oxidative metabolism of 5-methoxy-N,N-diisopropyltryptamine (Foxy) by human liver microsomes and recombinant cytochrome P450 enzymes. 5-methoxy-N,N-diisopropyltryptamine 24-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 16607661-2 2006 We describe the use of the dynamic light-scattering technique (DLS) to determine the stoichiometry of the protein cytochrome P450(BSbeta) attached to CdS and CdSe quantum dots (QDs). Cadmium 150-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-137 16607661-2 2006 We describe the use of the dynamic light-scattering technique (DLS) to determine the stoichiometry of the protein cytochrome P450(BSbeta) attached to CdS and CdSe quantum dots (QDs). cdse 158-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-137 16467135-1 2006 The objective of this study was to evaluate the duration of oral pleconaril (a picornavirus inhibitor) effect on intestinal and hepatic cytochrome P450 (P450) 3A activity as assessed by oral midazolam. pleconaril 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-161 16365879-7 2006 Furthermore, the UVB-dependent induction of 24-hydroxylase was blocked by the cytochrome-P450 inhibitor ketoconazole. Ketoconazole 104-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 16527260-0 2006 Cytochrome P450 expression and metabolic activation of cooked food mutagen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) in MCF10A breast epithelial cells. 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine 75-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 16527260-0 2006 Cytochrome P450 expression and metabolic activation of cooked food mutagen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) in MCF10A breast epithelial cells. 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine 124-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 16750462-7 2006 In vitro studies have indicated that the cytochrome P450 (CYP) isozymes CYP3A4 and CYP2E1 are involved in the biotransformation of eszopiclone; therefore, drugs that induce or inhibit these CYP isozymes may affect the metabolism of eszopiclone. Eszopiclone 131-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 16318905-0 2006 Characterization of human liver cytochrome P450 enzymes involved in the metabolism of rutaecarpine. rutecarpine 86-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 16318905-3 2006 The specific cytochrome P450 (CYP) isozymes responsible for rutaecarpine metabolites were identified using the combination of chemical inhibition, immuno-inhibition and metabolism by cDNA expressed CYP enzymes. rutecarpine 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 16318905-3 2006 The specific cytochrome P450 (CYP) isozymes responsible for rutaecarpine metabolites were identified using the combination of chemical inhibition, immuno-inhibition and metabolism by cDNA expressed CYP enzymes. rutecarpine 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 16318905-3 2006 The specific cytochrome P450 (CYP) isozymes responsible for rutaecarpine metabolites were identified using the combination of chemical inhibition, immuno-inhibition and metabolism by cDNA expressed CYP enzymes. rutecarpine 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-201 16750462-7 2006 In vitro studies have indicated that the cytochrome P450 (CYP) isozymes CYP3A4 and CYP2E1 are involved in the biotransformation of eszopiclone; therefore, drugs that induce or inhibit these CYP isozymes may affect the metabolism of eszopiclone. Eszopiclone 131-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 16750462-7 2006 In vitro studies have indicated that the cytochrome P450 (CYP) isozymes CYP3A4 and CYP2E1 are involved in the biotransformation of eszopiclone; therefore, drugs that induce or inhibit these CYP isozymes may affect the metabolism of eszopiclone. Eszopiclone 131-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 16750462-7 2006 In vitro studies have indicated that the cytochrome P450 (CYP) isozymes CYP3A4 and CYP2E1 are involved in the biotransformation of eszopiclone; therefore, drugs that induce or inhibit these CYP isozymes may affect the metabolism of eszopiclone. Eszopiclone 232-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 16750462-7 2006 In vitro studies have indicated that the cytochrome P450 (CYP) isozymes CYP3A4 and CYP2E1 are involved in the biotransformation of eszopiclone; therefore, drugs that induce or inhibit these CYP isozymes may affect the metabolism of eszopiclone. Eszopiclone 232-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 16750462-7 2006 In vitro studies have indicated that the cytochrome P450 (CYP) isozymes CYP3A4 and CYP2E1 are involved in the biotransformation of eszopiclone; therefore, drugs that induce or inhibit these CYP isozymes may affect the metabolism of eszopiclone. Eszopiclone 232-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 16525816-0 2006 Investigation of sarizotan"s impact on the pharmacokinetics of probe drugs for major cytochrome P450 isoenzymes: a combined cocktail trial. sarizotan 17-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 16609933-0 2006 Capillary electrophoretic investigation of the enantioselective metabolism of propafenone by human cytochrome P-450 SUPERSOMES: Evidence for atypical kinetics by CYP2D6 and CYP3A4. Propafenone 78-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 16490839-1 2006 Epoxyeicosatrienoic acids (EETs) are epoxides of arachidonic acid generated by cytochrome P450 (CYP) epoxygenases. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 16490839-1 2006 Epoxyeicosatrienoic acids (EETs) are epoxides of arachidonic acid generated by cytochrome P450 (CYP) epoxygenases. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 16490839-1 2006 Epoxyeicosatrienoic acids (EETs) are epoxides of arachidonic acid generated by cytochrome P450 (CYP) epoxygenases. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 16490839-1 2006 Epoxyeicosatrienoic acids (EETs) are epoxides of arachidonic acid generated by cytochrome P450 (CYP) epoxygenases. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 16490839-1 2006 Epoxyeicosatrienoic acids (EETs) are epoxides of arachidonic acid generated by cytochrome P450 (CYP) epoxygenases. Epoxy Compounds 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 16490839-1 2006 Epoxyeicosatrienoic acids (EETs) are epoxides of arachidonic acid generated by cytochrome P450 (CYP) epoxygenases. Epoxy Compounds 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 16490839-1 2006 Epoxyeicosatrienoic acids (EETs) are epoxides of arachidonic acid generated by cytochrome P450 (CYP) epoxygenases. Arachidonic Acid 49-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 16490839-1 2006 Epoxyeicosatrienoic acids (EETs) are epoxides of arachidonic acid generated by cytochrome P450 (CYP) epoxygenases. Arachidonic Acid 49-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 16490839-2 2006 The activation of CYP epoxygenases in endothelial cells is an important step in the NO and prostacyclin-independent vasodilatation of several vascular beds, and EETs have been identified as an endothelium-derived hyperpolarizing factor. Epoprostenol 91-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 16443667-0 2006 Identification of human liver cytochrome P450 enzymes responsible for the metabolism of lonafarnib (Sarasar). lonafarnib 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 16525816-2 2006 We investigated the effect of sarizotan on the clinical pharmacokinetics of probe drugs for cytochrome P450 (CYP) to evaluate the risk of CYP-related drug-drug interactions. sarizotan 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 16525816-2 2006 We investigated the effect of sarizotan on the clinical pharmacokinetics of probe drugs for cytochrome P450 (CYP) to evaluate the risk of CYP-related drug-drug interactions. sarizotan 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-112 16497287-3 2006 We showed that beta-carotene at 20 microM significantly enhanced NNK-induced DNA strand breaks and 7-mGua levels by 90% (p < 0.05) and 70% (p < 0.05), respectively, and that the effect of beta-carotene was associated with an increased metabolism of NNK by CYP because the concomitant addition of 1-aminobenzotriazole, a CYP inhibitor, with beta-carotene to cells strongly inhibited NNK-induced DNA strand breaks. beta Carotene 15-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-265 16483564-0 2006 Vitamins C and E protect hepatic cytochrome P450 dysfunction induced by polymicrobial sepsis. Carbon 9-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 16497287-3 2006 We showed that beta-carotene at 20 microM significantly enhanced NNK-induced DNA strand breaks and 7-mGua levels by 90% (p < 0.05) and 70% (p < 0.05), respectively, and that the effect of beta-carotene was associated with an increased metabolism of NNK by CYP because the concomitant addition of 1-aminobenzotriazole, a CYP inhibitor, with beta-carotene to cells strongly inhibited NNK-induced DNA strand breaks. beta Carotene 15-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 326-329 16251477-1 2006 Arachidonic acid (AA) is a substrate for a variety of proinflammatory mediators, which are generated by cyclooxygenases (COXs), lipoxygenases (LOXs), and cytochrome P-450 (CYP450) enzymes. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-170 16251477-1 2006 Arachidonic acid (AA) is a substrate for a variety of proinflammatory mediators, which are generated by cyclooxygenases (COXs), lipoxygenases (LOXs), and cytochrome P-450 (CYP450) enzymes. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-178 16251477-3 2006 We previously found that mechanical strain generated by subjecting lymphocytes to hypotonic challenge triggered AA production and that two CYP450 products of AA, 5,6-epoxyeicosatrienoic acid (5,6-EET) and 20-hydroxyeicosatetraenoic acid (20-HETE), as well as a product of LOX, 5-(S)-hydroperoxyeicosatetrenoic acid (5-HPETE), induced Ca(2+) entry into primary B cells. 5,6-epoxy-8,11,14-eicosatrienoic acid 162-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-145 16251477-3 2006 We previously found that mechanical strain generated by subjecting lymphocytes to hypotonic challenge triggered AA production and that two CYP450 products of AA, 5,6-epoxyeicosatrienoic acid (5,6-EET) and 20-hydroxyeicosatetraenoic acid (20-HETE), as well as a product of LOX, 5-(S)-hydroperoxyeicosatetrenoic acid (5-HPETE), induced Ca(2+) entry into primary B cells. 5,6-epoxy-8,11,14-eicosatrienoic acid 192-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-145 16251477-3 2006 We previously found that mechanical strain generated by subjecting lymphocytes to hypotonic challenge triggered AA production and that two CYP450 products of AA, 5,6-epoxyeicosatrienoic acid (5,6-EET) and 20-hydroxyeicosatetraenoic acid (20-HETE), as well as a product of LOX, 5-(S)-hydroperoxyeicosatetrenoic acid (5-HPETE), induced Ca(2+) entry into primary B cells. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 205-236 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-145 16251477-3 2006 We previously found that mechanical strain generated by subjecting lymphocytes to hypotonic challenge triggered AA production and that two CYP450 products of AA, 5,6-epoxyeicosatrienoic acid (5,6-EET) and 20-hydroxyeicosatetraenoic acid (20-HETE), as well as a product of LOX, 5-(S)-hydroperoxyeicosatetrenoic acid (5-HPETE), induced Ca(2+) entry into primary B cells. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 238-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-145 16251477-3 2006 We previously found that mechanical strain generated by subjecting lymphocytes to hypotonic challenge triggered AA production and that two CYP450 products of AA, 5,6-epoxyeicosatrienoic acid (5,6-EET) and 20-hydroxyeicosatetraenoic acid (20-HETE), as well as a product of LOX, 5-(S)-hydroperoxyeicosatetrenoic acid (5-HPETE), induced Ca(2+) entry into primary B cells. 5-(s)-hydroperoxyeicosatetrenoic acid 277-314 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-145 16251477-3 2006 We previously found that mechanical strain generated by subjecting lymphocytes to hypotonic challenge triggered AA production and that two CYP450 products of AA, 5,6-epoxyeicosatrienoic acid (5,6-EET) and 20-hydroxyeicosatetraenoic acid (20-HETE), as well as a product of LOX, 5-(S)-hydroperoxyeicosatetrenoic acid (5-HPETE), induced Ca(2+) entry into primary B cells. arachidonic acid 5-hydroperoxide 316-323 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-145 16487217-4 2006 Carbamazepine, phenytoin, phenobarbital and primidone induce many cytochrome P450 (CYP) and glucuronyl transferase (GT) enzymes, and can reduce drastically the serum concentration of associated drugs which are substrates of the same enzymes. Carbamazepine 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 16487217-4 2006 Carbamazepine, phenytoin, phenobarbital and primidone induce many cytochrome P450 (CYP) and glucuronyl transferase (GT) enzymes, and can reduce drastically the serum concentration of associated drugs which are substrates of the same enzymes. Carbamazepine 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 16487217-4 2006 Carbamazepine, phenytoin, phenobarbital and primidone induce many cytochrome P450 (CYP) and glucuronyl transferase (GT) enzymes, and can reduce drastically the serum concentration of associated drugs which are substrates of the same enzymes. Primidone 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 16487217-4 2006 Carbamazepine, phenytoin, phenobarbital and primidone induce many cytochrome P450 (CYP) and glucuronyl transferase (GT) enzymes, and can reduce drastically the serum concentration of associated drugs which are substrates of the same enzymes. Phenytoin 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 16487217-4 2006 Carbamazepine, phenytoin, phenobarbital and primidone induce many cytochrome P450 (CYP) and glucuronyl transferase (GT) enzymes, and can reduce drastically the serum concentration of associated drugs which are substrates of the same enzymes. Phenytoin 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 16487217-4 2006 Carbamazepine, phenytoin, phenobarbital and primidone induce many cytochrome P450 (CYP) and glucuronyl transferase (GT) enzymes, and can reduce drastically the serum concentration of associated drugs which are substrates of the same enzymes. Phenobarbital 26-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 16487217-4 2006 Carbamazepine, phenytoin, phenobarbital and primidone induce many cytochrome P450 (CYP) and glucuronyl transferase (GT) enzymes, and can reduce drastically the serum concentration of associated drugs which are substrates of the same enzymes. Phenobarbital 26-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 16487217-4 2006 Carbamazepine, phenytoin, phenobarbital and primidone induce many cytochrome P450 (CYP) and glucuronyl transferase (GT) enzymes, and can reduce drastically the serum concentration of associated drugs which are substrates of the same enzymes. Primidone 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 16436457-4 2006 FLJ39501 encodes a protein which was found to be a cytochrome P450, family 4, subfamily F, polypeptide 2 homolog of the leukotriene B4-omega-hydroxylase (CYP4F2) and could catalyze the 20-hydroxylation of trioxilin A3 from the 12(R)-lipoxygenase pathway. flj39501 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-152 16537715-4 2006 Therefore, we assessed the human cytochrome P450-dependent oxidation of estrone using substrate concentrations that more closely approximate the maximum expected concentrations in breast tissue. Estrone 72-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 16537715-5 2006 The in vitro metabolism of estrone by recombinant human cytochrome P450 enzymes and human liver microsomes was studied. Estrone 27-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 16436457-4 2006 FLJ39501 encodes a protein which was found to be a cytochrome P450, family 4, subfamily F, polypeptide 2 homolog of the leukotriene B4-omega-hydroxylase (CYP4F2) and could catalyze the 20-hydroxylation of trioxilin A3 from the 12(R)-lipoxygenase pathway. 8,11,12-trihydroxy-5,9,14-eicosatrienoic acid 205-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-152 16470567-10 2006 Imatinib metabolites were produced in microsomal incubations containing CYP isozymes. Imatinib Mesylate 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 16484569-1 2006 Eight water-soluble components of aged garlic extract were evaluated to assess their potential to inhibit the activity of human cytochrome-P450 (CYP) enzymes. Water 6-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-143 16484569-1 2006 Eight water-soluble components of aged garlic extract were evaluated to assess their potential to inhibit the activity of human cytochrome-P450 (CYP) enzymes. Water 6-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 16157466-1 2006 The oxidative metabolism of coumarin via several human cytochrome P450 (CYP) enzymes from families CYP1, CYP2 and CYP3 is rationalized in terms of molecular modelling studies carried out on the key interactions with various amino acid residues in the relevant active sites. coumarin 28-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 16289744-5 2006 This review focuses on the flavonoid effects on cytochrome P450 (CYP) enzymes involved in the activation of procarcinogens and phase II enzymes, largely responsible for the detoxification of carcinogens. Flavonoids 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 16289744-5 2006 This review focuses on the flavonoid effects on cytochrome P450 (CYP) enzymes involved in the activation of procarcinogens and phase II enzymes, largely responsible for the detoxification of carcinogens. Flavonoids 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 16157466-1 2006 The oxidative metabolism of coumarin via several human cytochrome P450 (CYP) enzymes from families CYP1, CYP2 and CYP3 is rationalized in terms of molecular modelling studies carried out on the key interactions with various amino acid residues in the relevant active sites. coumarin 28-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 16289744-6 2006 A number of naturally occurring flavonoids have been shown to modulate the CYP450 system, including the induction of specific CYP isozymes, and the activation or inhibition of these enzymes. Flavonoids 32-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 16719544-2 2006 In an attempt to explain this variability, three pharmacokinetic studies were carried out in healthy volunteers to investigate the relationship between exposure to gefitinib and cytochrome P450 (CYP) 3A phenotype (study 1), CYP3A5 genotype (study 2) and CYP2D6 genotype (study 3). Gefitinib 164-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 178-193 16401082-0 2006 Radical intermediates in the catalytic oxidation of hydrocarbons by bacterial and human cytochrome P450 enzymes. radical 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 16401082-0 2006 Radical intermediates in the catalytic oxidation of hydrocarbons by bacterial and human cytochrome P450 enzymes. Hydrocarbons 52-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 16280384-0 2006 Aflatoxin B1 alters the expression of p53 in cytochrome P450-expressing human lung cells. Aflatoxin B1 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 16719544-0 2006 Exploring the relationship between expression of cytochrome P450 enzymes and gefitinib pharmacokinetics. Gefitinib 77-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 16719544-2 2006 In an attempt to explain this variability, three pharmacokinetic studies were carried out in healthy volunteers to investigate the relationship between exposure to gefitinib and cytochrome P450 (CYP) 3A phenotype (study 1), CYP3A5 genotype (study 2) and CYP2D6 genotype (study 3). Gefitinib 164-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-198 16802848-10 2006 Clearance is hepatic via N-oxidation by the hepatic cytochrome P450 (CYP) isoenzymes, CYP2C19, CYP2C9 and CYP3A4. Nitrogen 25-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 16802848-10 2006 Clearance is hepatic via N-oxidation by the hepatic cytochrome P450 (CYP) isoenzymes, CYP2C19, CYP2C9 and CYP3A4. Nitrogen 25-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 16536934-1 2006 OBJECTIVES: To assess the relationship between the genetic and phenotypic factors linked to the cytochrome P-450 enzyme system and the response to the antimalarial drugs chloroquine, amodiaquine, mefloquine, and proguanil, as well as to determine how certain biological and social factors of the host influence the behavior of this enzymatic complex. Chloroquine 170-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 17181373-5 2006 For example, protease inhibitors often must be avoided if the potent CYP inducer rifampicin is co-administered. Rifampin 81-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 16379042-0 2006 In vitro characterization of the human biotransformation and CYP reaction phenotype of ET-743 (Yondelis, Trabectidin), a novel marine anti-cancer drug. Trabectedin 87-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 16379042-0 2006 In vitro characterization of the human biotransformation and CYP reaction phenotype of ET-743 (Yondelis, Trabectidin), a novel marine anti-cancer drug. trabectidin 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 16379042-2 2006 To assess the biotransformation and CYP reaction phenotype and their potential implications for human pharmacology and toxicology, the in vitro metabolism of ET-743 was characterized using incubations with human liver preparations, cytochrome P450 (CYP) and uridine diphosphoglucuronosyl transferase (UGT) supersomes.CYP supersomes and liver microsomes showed that ET-743 was metabolized mainly by CYP3A4, but also by CYP2C9, 2C19, 2D6, and 2E1. Trabectedin 158-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 16379042-2 2006 To assess the biotransformation and CYP reaction phenotype and their potential implications for human pharmacology and toxicology, the in vitro metabolism of ET-743 was characterized using incubations with human liver preparations, cytochrome P450 (CYP) and uridine diphosphoglucuronosyl transferase (UGT) supersomes.CYP supersomes and liver microsomes showed that ET-743 was metabolized mainly by CYP3A4, but also by CYP2C9, 2C19, 2D6, and 2E1. Trabectedin 158-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-247 16379042-2 2006 To assess the biotransformation and CYP reaction phenotype and their potential implications for human pharmacology and toxicology, the in vitro metabolism of ET-743 was characterized using incubations with human liver preparations, cytochrome P450 (CYP) and uridine diphosphoglucuronosyl transferase (UGT) supersomes.CYP supersomes and liver microsomes showed that ET-743 was metabolized mainly by CYP3A4, but also by CYP2C9, 2C19, 2D6, and 2E1. Trabectedin 158-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 249-252 16379042-2 2006 To assess the biotransformation and CYP reaction phenotype and their potential implications for human pharmacology and toxicology, the in vitro metabolism of ET-743 was characterized using incubations with human liver preparations, cytochrome P450 (CYP) and uridine diphosphoglucuronosyl transferase (UGT) supersomes.CYP supersomes and liver microsomes showed that ET-743 was metabolized mainly by CYP3A4, but also by CYP2C9, 2C19, 2D6, and 2E1. Trabectedin 158-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 249-252 16379042-7 2006 ET-743 was more extensively metabolized when CYP activity was combined with phase II enzymes UGT and glutathione-S-transferase (GST), indicating that CYP, UGT, and GST simultaneously metabolize ET-743 in the S9 fraction. Trabectedin 0-6 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 16379042-7 2006 ET-743 was more extensively metabolized when CYP activity was combined with phase II enzymes UGT and glutathione-S-transferase (GST), indicating that CYP, UGT, and GST simultaneously metabolize ET-743 in the S9 fraction. Trabectedin 0-6 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 16379042-7 2006 ET-743 was more extensively metabolized when CYP activity was combined with phase II enzymes UGT and glutathione-S-transferase (GST), indicating that CYP, UGT, and GST simultaneously metabolize ET-743 in the S9 fraction. Trabectedin 194-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 16379042-7 2006 ET-743 was more extensively metabolized when CYP activity was combined with phase II enzymes UGT and glutathione-S-transferase (GST), indicating that CYP, UGT, and GST simultaneously metabolize ET-743 in the S9 fraction. Trabectedin 194-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 16719370-4 2006 CYP are b-type cytochromes, containing protoporphyrin IX as the prosthetic group. protoporphyrin IX 39-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 16719370-6 2006 In its oxidized, or ferric state, CYP exists as an equilibrium mixture of high- and low-spin configurations, each with distinctive UV/Vis absorption peaks. Ferric enterobactin ion 20-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 16497109-2 2006 Systemic blood pressure, after inhibitory intervention in arachidonic acid metabolism cytochrome P-450 inhibition by miconazole 0.5 mg/100 g b.w. Arachidonic Acid 58-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 16497109-2 2006 Systemic blood pressure, after inhibitory intervention in arachidonic acid metabolism cytochrome P-450 inhibition by miconazole 0.5 mg/100 g b.w. Miconazole 117-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 16944963-5 2006 Proton pump inhibitors also influence drug absorption and metabolism by interacting with adenosine triphosphate-dependent P-glycoprotein (e.g. inhibiting digoxin efflux) or with the cytochrome P450 (CYP) enzyme system (e.g. decreasing simvastatin metabolism), thereby affecting both intestinal first-pass metabolism and hepatic clearance. Simvastatin 235-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-197 16944963-5 2006 Proton pump inhibitors also influence drug absorption and metabolism by interacting with adenosine triphosphate-dependent P-glycoprotein (e.g. inhibiting digoxin efflux) or with the cytochrome P450 (CYP) enzyme system (e.g. decreasing simvastatin metabolism), thereby affecting both intestinal first-pass metabolism and hepatic clearance. Simvastatin 235-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 199-202 16338087-2 2006 The first step in steroidogenesis involves the delivery of free cholesterol to the inner mitochondrial membrane where it can be converted into pregnenolone by the enzyme cytochrome P450side chain cleavage. Cholesterol 64-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-185 16338087-2 2006 The first step in steroidogenesis involves the delivery of free cholesterol to the inner mitochondrial membrane where it can be converted into pregnenolone by the enzyme cytochrome P450side chain cleavage. Pregnenolone 143-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-185 16541400-0 2006 Bilirubin degradation by uncoupled cytochrome P450. Bilirubin 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 16541400-3 2006 Degradation of bilirubin takes place (a) on interaction with oxidative free radicals and (b) when cytochrome P450 (CYP) enzymes are uncoupled by polyhalogenated substrate analogues. Bilirubin 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 16541400-3 2006 Degradation of bilirubin takes place (a) on interaction with oxidative free radicals and (b) when cytochrome P450 (CYP) enzymes are uncoupled by polyhalogenated substrate analogues. Bilirubin 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 16541400-10 2006 Fe-EDTA/H2O2 and uncoupled CYP(Fe=O) may both initiate the reaction, the latter in an attempt to reduce the ferryl oxygen to water. Oxygen 115-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 16541400-10 2006 Fe-EDTA/H2O2 and uncoupled CYP(Fe=O) may both initiate the reaction, the latter in an attempt to reduce the ferryl oxygen to water. Water 125-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 16536934-1 2006 OBJECTIVES: To assess the relationship between the genetic and phenotypic factors linked to the cytochrome P-450 enzyme system and the response to the antimalarial drugs chloroquine, amodiaquine, mefloquine, and proguanil, as well as to determine how certain biological and social factors of the host influence the behavior of this enzymatic complex. Amodiaquine 183-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 16536934-1 2006 OBJECTIVES: To assess the relationship between the genetic and phenotypic factors linked to the cytochrome P-450 enzyme system and the response to the antimalarial drugs chloroquine, amodiaquine, mefloquine, and proguanil, as well as to determine how certain biological and social factors of the host influence the behavior of this enzymatic complex. Proguanil 212-221 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 16143297-0 2005 Steroid hydroxylations: a paradigm for cytochrome P450 catalyzed mammalian monooxygenation reactions. Steroids 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 16183271-0 2005 A thiolate ligand on a cytochrome P-450 mimic permits the use of simple environmentally benign oxidants for biomimetic steroid hydroxylation in water. thiolate 2-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 16183271-0 2005 A thiolate ligand on a cytochrome P-450 mimic permits the use of simple environmentally benign oxidants for biomimetic steroid hydroxylation in water. Steroids 119-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 16183271-0 2005 A thiolate ligand on a cytochrome P-450 mimic permits the use of simple environmentally benign oxidants for biomimetic steroid hydroxylation in water. Water 144-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 16137656-0 2005 First evidence that cytochrome P450 may catalyze both S-oxidation and epoxidation of thiophene derivatives. Thiophenes 85-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 16137656-5 2005 These results provide the first evidence that cytochrome P450 may catalyze the oxidation of thiophene compounds with the simultaneous formation of two reactive intermediates, a thiophene-S-oxide and a thiophene epoxide. Thiophenes 92-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 16375696-1 2005 Cytochrome P450 (CYP) is a group of enzymes that metabolize drugs to a more water-soluble form, rendering them available for renal excretion. Water 76-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 16137656-5 2005 These results provide the first evidence that cytochrome P450 may catalyze the oxidation of thiophene compounds with the simultaneous formation of two reactive intermediates, a thiophene-S-oxide and a thiophene epoxide. thiophene-s-oxide 177-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 16137656-5 2005 These results provide the first evidence that cytochrome P450 may catalyze the oxidation of thiophene compounds with the simultaneous formation of two reactive intermediates, a thiophene-S-oxide and a thiophene epoxide. thiophene epoxide 201-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 16207711-1 2005 Cytochrome P450 (P450) 2A6 is an important human enzyme involved in the metabolism of many xenobiotic chemicals including coumarin, indole, nicotine, and carcinogenic nitrosamines. coumarin 122-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 16207711-1 2005 Cytochrome P450 (P450) 2A6 is an important human enzyme involved in the metabolism of many xenobiotic chemicals including coumarin, indole, nicotine, and carcinogenic nitrosamines. indole 132-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 16207711-1 2005 Cytochrome P450 (P450) 2A6 is an important human enzyme involved in the metabolism of many xenobiotic chemicals including coumarin, indole, nicotine, and carcinogenic nitrosamines. Nicotine 140-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 16207711-1 2005 Cytochrome P450 (P450) 2A6 is an important human enzyme involved in the metabolism of many xenobiotic chemicals including coumarin, indole, nicotine, and carcinogenic nitrosamines. Nitrosamines 167-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 16126163-5 2005 Oxy-I resembles the ferrous oxygen complex known for cytochrome P450, whereas oxy-II appears to be locked in the superoxide form. oxy-i 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-84 16126163-5 2005 Oxy-I resembles the ferrous oxygen complex known for cytochrome P450, whereas oxy-II appears to be locked in the superoxide form. ferrous oxygen 20-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-84 16126163-5 2005 Oxy-I resembles the ferrous oxygen complex known for cytochrome P450, whereas oxy-II appears to be locked in the superoxide form. Superoxides 113-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-84 16322314-5 2005 Studies with cDNA-expressed human cytochrome P450 enzymes revealed that BPU was metabolized predominantly by CYP3A4 and CYP1A1 but was also a substrate for CYP2C8, CYP2D6, CYP3A5, and CYP3A7. bpu 72-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 16322314-12 2005 Thus, we conclude that (a) CYP3A4 and CYP1A1 are the predominant cytochrome P450 enzymes that catalyze BPU metabolism, (b) BPU is metabolized to two cytotoxic and four noncytotoxic metabolites, and (c) ritonavir inhibits BPU metabolism to improve the systemic exposure to BPU without altering cumulative exposure to BPU and the cytotoxic metabolites. bpu 103-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 16322314-12 2005 Thus, we conclude that (a) CYP3A4 and CYP1A1 are the predominant cytochrome P450 enzymes that catalyze BPU metabolism, (b) BPU is metabolized to two cytotoxic and four noncytotoxic metabolites, and (c) ritonavir inhibits BPU metabolism to improve the systemic exposure to BPU without altering cumulative exposure to BPU and the cytotoxic metabolites. Ritonavir 202-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 16322314-12 2005 Thus, we conclude that (a) CYP3A4 and CYP1A1 are the predominant cytochrome P450 enzymes that catalyze BPU metabolism, (b) BPU is metabolized to two cytotoxic and four noncytotoxic metabolites, and (c) ritonavir inhibits BPU metabolism to improve the systemic exposure to BPU without altering cumulative exposure to BPU and the cytotoxic metabolites. bpu 123-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 16322314-12 2005 Thus, we conclude that (a) CYP3A4 and CYP1A1 are the predominant cytochrome P450 enzymes that catalyze BPU metabolism, (b) BPU is metabolized to two cytotoxic and four noncytotoxic metabolites, and (c) ritonavir inhibits BPU metabolism to improve the systemic exposure to BPU without altering cumulative exposure to BPU and the cytotoxic metabolites. bpu 123-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 16322314-12 2005 Thus, we conclude that (a) CYP3A4 and CYP1A1 are the predominant cytochrome P450 enzymes that catalyze BPU metabolism, (b) BPU is metabolized to two cytotoxic and four noncytotoxic metabolites, and (c) ritonavir inhibits BPU metabolism to improve the systemic exposure to BPU without altering cumulative exposure to BPU and the cytotoxic metabolites. bpu 123-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 16375696-1 2005 Cytochrome P450 (CYP) is a group of enzymes that metabolize drugs to a more water-soluble form, rendering them available for renal excretion. Water 76-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 16202979-2 2005 It has been reported that the metabolic activation of PAHs by cytochrome P450 (CYP) is an important step for PAH-induced atherosclerosis. Polycyclic Aromatic Hydrocarbons 54-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 16291720-1 2005 Recent studies suggest that cytochrome P450 (CYP) epoxygenase-derived epoxyeicosatrienoic acids (EETs) elicit cell proliferation and promote angiogenesis. epoxyeicosatrienoic acids 70-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 16291720-1 2005 Recent studies suggest that cytochrome P450 (CYP) epoxygenase-derived epoxyeicosatrienoic acids (EETs) elicit cell proliferation and promote angiogenesis. epoxyeicosatrienoic acids 70-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 16291720-1 2005 Recent studies suggest that cytochrome P450 (CYP) epoxygenase-derived epoxyeicosatrienoic acids (EETs) elicit cell proliferation and promote angiogenesis. eets 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 16291720-1 2005 Recent studies suggest that cytochrome P450 (CYP) epoxygenase-derived epoxyeicosatrienoic acids (EETs) elicit cell proliferation and promote angiogenesis. eets 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 16291720-4 2005 In Transwell assays, the migration of endothelial cells pre-exposed to hypoxia to increase CYP expression was abolished by CYP 2C antisense oligonucleotides as well as by the CYP inhibitor MS-PPOH and the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (EEZE). Oligonucleotides 140-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 16291720-4 2005 In Transwell assays, the migration of endothelial cells pre-exposed to hypoxia to increase CYP expression was abolished by CYP 2C antisense oligonucleotides as well as by the CYP inhibitor MS-PPOH and the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (EEZE). Oligonucleotides 140-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 16291720-4 2005 In Transwell assays, the migration of endothelial cells pre-exposed to hypoxia to increase CYP expression was abolished by CYP 2C antisense oligonucleotides as well as by the CYP inhibitor MS-PPOH and the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (EEZE). N-methylsulfonyl-6-(2-propargyloxyphenyl)hexanamide 189-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 16291720-4 2005 In Transwell assays, the migration of endothelial cells pre-exposed to hypoxia to increase CYP expression was abolished by CYP 2C antisense oligonucleotides as well as by the CYP inhibitor MS-PPOH and the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (EEZE). 14,15-episulfide eicosatrienoic acid 220-253 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 16291720-4 2005 In Transwell assays, the migration of endothelial cells pre-exposed to hypoxia to increase CYP expression was abolished by CYP 2C antisense oligonucleotides as well as by the CYP inhibitor MS-PPOH and the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (EEZE). 14,15-episulfide eicosatrienoic acid 220-253 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 18360572-6 2005 Itraconazole works by inhibiting ergosterol synthesis via cytochrome P-450 (CYP450)-dependent demethylation step. Itraconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 18360572-6 2005 Itraconazole works by inhibiting ergosterol synthesis via cytochrome P-450 (CYP450)-dependent demethylation step. Itraconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-82 18360572-6 2005 Itraconazole works by inhibiting ergosterol synthesis via cytochrome P-450 (CYP450)-dependent demethylation step. Ergosterol 33-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 18360572-6 2005 Itraconazole works by inhibiting ergosterol synthesis via cytochrome P-450 (CYP450)-dependent demethylation step. Ergosterol 33-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-82 16202979-2 2005 It has been reported that the metabolic activation of PAHs by cytochrome P450 (CYP) is an important step for PAH-induced atherosclerosis. Polycyclic Aromatic Hydrocarbons 54-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 16202979-2 2005 It has been reported that the metabolic activation of PAHs by cytochrome P450 (CYP) is an important step for PAH-induced atherosclerosis. Polycyclic Aromatic Hydrocarbons 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 16202979-2 2005 It has been reported that the metabolic activation of PAHs by cytochrome P450 (CYP) is an important step for PAH-induced atherosclerosis. Polycyclic Aromatic Hydrocarbons 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 16243813-0 2005 Effect of common CYP3A4 and CYP3A5 variants on the pharmacokinetics of the cytochrome P450 3A phenotyping probe midazolam in cancer patients. Midazolam 112-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 16236141-3 2005 In vitro and in vivo data suggest that the metabolism of fluoxetine to norfluoxetine is stereoselective and mediated, at least in part, by the polymorphic cytochrome P450 (CYP) isoenzymes CYP2D6, CYP2C9 and CYP2C19. Fluoxetine 57-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 16236141-3 2005 In vitro and in vivo data suggest that the metabolism of fluoxetine to norfluoxetine is stereoselective and mediated, at least in part, by the polymorphic cytochrome P450 (CYP) isoenzymes CYP2D6, CYP2C9 and CYP2C19. Fluoxetine 57-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 16236141-3 2005 In vitro and in vivo data suggest that the metabolism of fluoxetine to norfluoxetine is stereoselective and mediated, at least in part, by the polymorphic cytochrome P450 (CYP) isoenzymes CYP2D6, CYP2C9 and CYP2C19. norfluoxetine 71-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 16236141-3 2005 In vitro and in vivo data suggest that the metabolism of fluoxetine to norfluoxetine is stereoselective and mediated, at least in part, by the polymorphic cytochrome P450 (CYP) isoenzymes CYP2D6, CYP2C9 and CYP2C19. norfluoxetine 71-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-175 16300378-0 2005 Bioactivation of dibrominated biphenyls by cytochrome P450 activity to metabolites with estrogenic activity and estrogen sulfotransferase inhibition capacity. diphenyl 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 16243813-1 2005 PURPOSE: To evaluate the effect of naturally occurring variants in genes encoding the cytochrome P450 (CYP) isoforms CYP3A4 and CYP3A5 in patients with cancer receiving midazolam as a phenotyping probe. Midazolam 169-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 16243813-1 2005 PURPOSE: To evaluate the effect of naturally occurring variants in genes encoding the cytochrome P450 (CYP) isoforms CYP3A4 and CYP3A5 in patients with cancer receiving midazolam as a phenotyping probe. Midazolam 169-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 16162970-3 2005 In-vitro studies have shown that ET-743 is mainly metabolized by cytochrome P450 (CYP) 3A4, but also by 2C9, 2C19, 2D6 and 2E1, and the phase II enzymes uridine diphosphoglucuronosyl transferase and glutathione-S-transferase. Trabectedin 33-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 16162970-3 2005 In-vitro studies have shown that ET-743 is mainly metabolized by cytochrome P450 (CYP) 3A4, but also by 2C9, 2C19, 2D6 and 2E1, and the phase II enzymes uridine diphosphoglucuronosyl transferase and glutathione-S-transferase. Trabectedin 33-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 16162970-5 2005 Therefore, the effect of CYP and phase II activity on the cytotoxicity of ET-743 was investigated in vitro in a human cell line model system. Trabectedin 74-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 16162970-6 2005 The effect of different CYP and phase II inhibitors and CYP inducers on ET-743 cytotoxicity was studied after 48 and 120 h of treatment in Hep G2 cells using different assays. Trabectedin 72-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 16162970-6 2005 The effect of different CYP and phase II inhibitors and CYP inducers on ET-743 cytotoxicity was studied after 48 and 120 h of treatment in Hep G2 cells using different assays. Trabectedin 72-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 16276979-4 2005 Cimetidine (CMT) inhibits the breakdown of drugs metabolized by CYP450 and reduces the clearance of coadministered drug resulted from both the CMT binding to CYP450 and the decreased hepatic blood flow due to CMT. Cimetidine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-70 16162970-8 2005 Potent cytotoxic activity of ET-743 after 120 h treatment was observed, which could be increased in combination with the CYP inhibitors metyrapone (3A4), phenanthrene (substrate for 2E1, 3A4), piperonyl butoxide (3A), proadifen (2C9, 2E1, 3A4), ritonavir (3A4), and warfarin (2C9, 2C19). Trabectedin 29-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 16276979-4 2005 Cimetidine (CMT) inhibits the breakdown of drugs metabolized by CYP450 and reduces the clearance of coadministered drug resulted from both the CMT binding to CYP450 and the decreased hepatic blood flow due to CMT. Cimetidine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-164 16276979-4 2005 Cimetidine (CMT) inhibits the breakdown of drugs metabolized by CYP450 and reduces the clearance of coadministered drug resulted from both the CMT binding to CYP450 and the decreased hepatic blood flow due to CMT. Cimetidine 12-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-70 16162970-8 2005 Potent cytotoxic activity of ET-743 after 120 h treatment was observed, which could be increased in combination with the CYP inhibitors metyrapone (3A4), phenanthrene (substrate for 2E1, 3A4), piperonyl butoxide (3A), proadifen (2C9, 2E1, 3A4), ritonavir (3A4), and warfarin (2C9, 2C19). Metyrapone 136-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 16276979-4 2005 Cimetidine (CMT) inhibits the breakdown of drugs metabolized by CYP450 and reduces the clearance of coadministered drug resulted from both the CMT binding to CYP450 and the decreased hepatic blood flow due to CMT. Cimetidine 12-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-164 16162970-8 2005 Potent cytotoxic activity of ET-743 after 120 h treatment was observed, which could be increased in combination with the CYP inhibitors metyrapone (3A4), phenanthrene (substrate for 2E1, 3A4), piperonyl butoxide (3A), proadifen (2C9, 2E1, 3A4), ritonavir (3A4), and warfarin (2C9, 2C19). phenanthrene 154-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 16276979-4 2005 Cimetidine (CMT) inhibits the breakdown of drugs metabolized by CYP450 and reduces the clearance of coadministered drug resulted from both the CMT binding to CYP450 and the decreased hepatic blood flow due to CMT. Cimetidine 143-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-164 16276979-4 2005 Cimetidine (CMT) inhibits the breakdown of drugs metabolized by CYP450 and reduces the clearance of coadministered drug resulted from both the CMT binding to CYP450 and the decreased hepatic blood flow due to CMT. Cimetidine 143-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-164 16162970-8 2005 Potent cytotoxic activity of ET-743 after 120 h treatment was observed, which could be increased in combination with the CYP inhibitors metyrapone (3A4), phenanthrene (substrate for 2E1, 3A4), piperonyl butoxide (3A), proadifen (2C9, 2E1, 3A4), ritonavir (3A4), and warfarin (2C9, 2C19). Piperonyl Butoxide 193-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 16162970-8 2005 Potent cytotoxic activity of ET-743 after 120 h treatment was observed, which could be increased in combination with the CYP inhibitors metyrapone (3A4), phenanthrene (substrate for 2E1, 3A4), piperonyl butoxide (3A), proadifen (2C9, 2E1, 3A4), ritonavir (3A4), and warfarin (2C9, 2C19). Proadifen 218-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 16162970-8 2005 Potent cytotoxic activity of ET-743 after 120 h treatment was observed, which could be increased in combination with the CYP inhibitors metyrapone (3A4), phenanthrene (substrate for 2E1, 3A4), piperonyl butoxide (3A), proadifen (2C9, 2E1, 3A4), ritonavir (3A4), and warfarin (2C9, 2C19). Ritonavir 245-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 16162970-8 2005 Potent cytotoxic activity of ET-743 after 120 h treatment was observed, which could be increased in combination with the CYP inhibitors metyrapone (3A4), phenanthrene (substrate for 2E1, 3A4), piperonyl butoxide (3A), proadifen (2C9, 2E1, 3A4), ritonavir (3A4), and warfarin (2C9, 2C19). Warfarin 266-274 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 16162970-10 2005 CYP metabolites of ET-743 were less toxic compared with ET-743. Trabectedin 19-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 16162970-11 2005 These findings indicate that combination therapy of ET-743 with CYP inhibitors, e.g. other anti-cancer drugs, could lead to changes in the hepatotoxicity of ET-743 and are therefore of clinical importance. Trabectedin 52-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 16162970-11 2005 These findings indicate that combination therapy of ET-743 with CYP inhibitors, e.g. other anti-cancer drugs, could lead to changes in the hepatotoxicity of ET-743 and are therefore of clinical importance. Trabectedin 157-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 15916533-0 2005 Cytochrome P450/NADPH-dependent biosynthesis of 5,6-trans-epoxyeicosatrienoic acid from 5,6-trans-arachidonic acid. 5,6-trans-epoxyeicosatrienoic acid 48-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-21 16408808-2 2005 METHODS: Imrecoxib was incubated with heterologous expression human cytochrome P450 (rCYPs) in vitro, and metabolites and remained parent drug were detected with liquid chromatography-multistage mass spectrometry. Imrecoxib 9-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 15993870-4 2005 Several lines of data support the possibility that certain CYP metabolites resulting from the hydroxylation of AA such as 20-hydroxyeicosatetraenoic acid (20-HETE) are potent vasoconstrictors and may produce detrimental effects in the heart during ischemia and pro-inflammatory effects during reperfusion. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 122-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 15993870-4 2005 Several lines of data support the possibility that certain CYP metabolites resulting from the hydroxylation of AA such as 20-hydroxyeicosatetraenoic acid (20-HETE) are potent vasoconstrictors and may produce detrimental effects in the heart during ischemia and pro-inflammatory effects during reperfusion. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 155-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 16298597-1 2005 Cytochrome-P450 enzymes metabolize cyclosporine both in the liver and in the intestinal wall. Cyclosporine 35-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 16051450-0 2005 Two novel transposable elements in a cytochrome P450 gene govern anthocyanin biosynthesis of commercial petunias. Anthocyanins 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 15916533-0 2005 Cytochrome P450/NADPH-dependent biosynthesis of 5,6-trans-epoxyeicosatrienoic acid from 5,6-trans-arachidonic acid. 5(E)-Arachidonic acid 88-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-21 16111645-8 2005 Depending on the examined system, this parallelism may be distorted due to the cytochrome P-450 and COMT-catalyzed transformation of polyphenols. Polyphenols 133-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 15993852-0 2005 Role of Ca2+-independent phospholipase A2 and cytochrome P-450 in store-operated calcium entry in 3T6 fibroblasts. Calcium 81-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 15993852-3 2005 Thapsigargin-induced elevation of [Ca2+]i was inhibited by cytochrome P-450 inhibitors and by cytochrome P-450 epoxygenase inhibitor and was reverted by 11,12 EET addition. Thapsigargin 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 15993852-3 2005 Thapsigargin-induced elevation of [Ca2+]i was inhibited by cytochrome P-450 inhibitors and by cytochrome P-450 epoxygenase inhibitor and was reverted by 11,12 EET addition. Thapsigargin 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 15993852-7 2005 Thus, AA released by Ca2+-independent phospholipase A2 and AA metabolism through cytochrome P-450 pathway may be crucial molecular determinant in thapsigargin activation of SOC channels and store-operated Ca2+ entry pathway in 3T6 fibroblasts. Thapsigargin 146-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 15993852-8 2005 Moreover, EETs, the main cytochrome P-450 epoxygenase metabolites of AA, are involved in thapsigargin-stimulated Ca2+ influx. Thapsigargin 89-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 16011979-1 2005 The antiplatelet potency of clopidogrel may be attenuated by short-term co-administration of lipophilic statins metabolized through the cytochrome P-450, isoform 3A4. Clopidogrel 28-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-152 15896668-6 2005 Results showed that treatment with CP-55,940 decreased CYP concentrations by 80% and increased HO activity by 158%. cp-55 35-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 16022919-1 2005 To clarify the role of cytochrome P450 in docetaxel and cisplatin combination chemotherapy, cytochrome P450 activity was measured by simple antipyrine test, and its correlation with the drugs" pharmacodynamics was assessed. Docetaxel 42-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 16022919-1 2005 To clarify the role of cytochrome P450 in docetaxel and cisplatin combination chemotherapy, cytochrome P450 activity was measured by simple antipyrine test, and its correlation with the drugs" pharmacodynamics was assessed. Cisplatin 56-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 16022919-9 2005 We concluded that antipyrine test and cytochrome P450 play an important role in predicting toxicities of docetaxel and cisplatin combination chemotherapy. Docetaxel 105-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 16022919-9 2005 We concluded that antipyrine test and cytochrome P450 play an important role in predicting toxicities of docetaxel and cisplatin combination chemotherapy. Cisplatin 119-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 16087391-0 2005 Nitrogen-substitution effects on the mutagenicity and cytochrome P450 isoform-selectivity of chrysene analogs. Nitrogen 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 16087391-0 2005 Nitrogen-substitution effects on the mutagenicity and cytochrome P450 isoform-selectivity of chrysene analogs. chrysene 93-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 16087391-8 2005 However, in Ames tests using microsomes from insect cells expressing various human CYP isoforms, the mutagenicity of 1,10-DAC was induced only by recombinant human CYP1A2, whereas both recombinant human CYP2A6 and 1A2 contributed to the mutagenicity of 4,10-DAC. 1,10-diazachrysene 117-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 16087391-8 2005 However, in Ames tests using microsomes from insect cells expressing various human CYP isoforms, the mutagenicity of 1,10-DAC was induced only by recombinant human CYP1A2, whereas both recombinant human CYP2A6 and 1A2 contributed to the mutagenicity of 4,10-DAC. 4,10-dac 253-261 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 16087391-10 2005 In conclusion, our results suggested that the difference in the nitrogen-substituted position in the chrysene molecule might affect the mutagenic activity through influencing the ratio of participation of the metabolic activation enzyme isoforms of CYP. Nitrogen 64-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 249-252 16087391-10 2005 In conclusion, our results suggested that the difference in the nitrogen-substituted position in the chrysene molecule might affect the mutagenic activity through influencing the ratio of participation of the metabolic activation enzyme isoforms of CYP. chrysene 101-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 249-252 16922637-4 2005 These interactions may occur not only in the liver, but also in the brain, and may change the activity of CYP towards the metabolism of drugs, sex steroids, neurosteroids and amine neurotransmitters. Steroids 147-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 16922637-4 2005 These interactions may occur not only in the liver, but also in the brain, and may change the activity of CYP towards the metabolism of drugs, sex steroids, neurosteroids and amine neurotransmitters. Amines 175-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 16278191-1 2005 The role of the major drug-metabolizing cytochrome P450 (CYP) enzymes as well as P-glycoprotein (PGP) was investigated in the disposition of ketobemidone in vitro. ketobemidone 141-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 16278191-1 2005 The role of the major drug-metabolizing cytochrome P450 (CYP) enzymes as well as P-glycoprotein (PGP) was investigated in the disposition of ketobemidone in vitro. ketobemidone 141-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 27722966-13 2005 In contrast, sulphonylureas, meglitinide derivatives and thiazolidinediones are extensively metabolised in the liver via the CYP system and thus, may be subject to drug-drug metabolic interactions. meglitinide 29-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 15963101-1 2005 AIMS: The genetically polymorphic cytochrome P450 (CYP) enzyme CYP2C9 metabolizes most sulphonylurea oral hypoglycaemic agents. Sulfonylurea Compounds 87-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 15963101-1 2005 AIMS: The genetically polymorphic cytochrome P450 (CYP) enzyme CYP2C9 metabolizes most sulphonylurea oral hypoglycaemic agents. Sulfonylurea Compounds 87-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 16003296-1 2005 The tuberculostatic compound rifampin (INN, rifampicin) induces the expression of a number of drug metabolism-related genes involved in multidrug resistance (P-glycoprotein and multidrug resistance proteins 1 and 2), cytochromes (cytochrome P450 [CYP] 3A4), uridine diphosphate-glucuronosyltransferases, monoamine oxidases, and glutathione S -transferases. Rifampin 29-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-255 16003296-1 2005 The tuberculostatic compound rifampin (INN, rifampicin) induces the expression of a number of drug metabolism-related genes involved in multidrug resistance (P-glycoprotein and multidrug resistance proteins 1 and 2), cytochromes (cytochrome P450 [CYP] 3A4), uridine diphosphate-glucuronosyltransferases, monoamine oxidases, and glutathione S -transferases. Rifampin 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-255 16178456-1 2005 Interests on the effects of cytochrome P450 (CYP450) monooxygenases and epoxyeicosatrienoic acids (EETs) on myocardial ischemic-reperfusion injury has been increased in recent years. eets 99-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-51 16178456-2 2005 The CYP450/EET system may influence the degree of myocardial ischemic-reperfusion injury through "poly-targets", such us oxygen free radical, calcium overload, leukocytes adherence, nitric oxide, ATP-sensitive K+ channels, and mitogen activated protein kinase. oxygen free radical 121-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-10 16178456-2 2005 The CYP450/EET system may influence the degree of myocardial ischemic-reperfusion injury through "poly-targets", such us oxygen free radical, calcium overload, leukocytes adherence, nitric oxide, ATP-sensitive K+ channels, and mitogen activated protein kinase. Calcium 142-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-10 27722966-13 2005 In contrast, sulphonylureas, meglitinide derivatives and thiazolidinediones are extensively metabolised in the liver via the CYP system and thus, may be subject to drug-drug metabolic interactions. Thiazolidinediones 57-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 16002382-2 2005 Organophosphorus insecticides (OPs) target the developing nervous system, and until recently, the most common residential insecticides were chlorpyrifos and diazinon, two OPs metabolized in the body through the cytochrome P450/paraoxonase 1 (PON1) pathway. organophosphorus 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-240 27722966-19 2005 The effects of inducers or inhibitors of CYP isoenzymes on the metabolism and pharmacokinetics of the glucose-lowering agents of each pharmacological class has been tested. Glucose 102-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 16002382-2 2005 Organophosphorus insecticides (OPs) target the developing nervous system, and until recently, the most common residential insecticides were chlorpyrifos and diazinon, two OPs metabolized in the body through the cytochrome P450/paraoxonase 1 (PON1) pathway. Chlorpyrifos 140-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-240 27722966-20 2005 Significantly increased (with CYP inhibitors) or decreased (with CYP inducers) plasma levels of sulphonylureas, meglitinide derivatives and thiazolidinediones have been reported in healthy volunteers, and these pharmacokinetic changes may lead to enhanced or reduced glucose-lowering action, and thus hypoglycaemia or worsening of metabolic control, respectively. meglitinide 112-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 16002382-2 2005 Organophosphorus insecticides (OPs) target the developing nervous system, and until recently, the most common residential insecticides were chlorpyrifos and diazinon, two OPs metabolized in the body through the cytochrome P450/paraoxonase 1 (PON1) pathway. Diazinon 157-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-240 27722966-20 2005 Significantly increased (with CYP inhibitors) or decreased (with CYP inducers) plasma levels of sulphonylureas, meglitinide derivatives and thiazolidinediones have been reported in healthy volunteers, and these pharmacokinetic changes may lead to enhanced or reduced glucose-lowering action, and thus hypoglycaemia or worsening of metabolic control, respectively. Thiazolidinediones 140-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 16025433-2 2005 Chloroform targets the liver in humans, where cytochrome P-450-dependent biotransformation to phosgene results in mitochondrial damage and cell death. Chloroform 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 15931672-0 2005 Arachidonic acid induces neuronal death through lipoxygenase and cytochrome P450 rather than cyclooxygenase. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 15931672-4 2005 Exposure of cultured cortical neurons to AA (50 microM) yielded significantly apoptotic neuronal death, which was attenuated greatly by LOX inhibitors (nordihydroguaiaretic acid, AA861, and baicalein), or CYP450 inhibitors (SKF525A and metyrapone), rather than COX inhibitors (indomethacin and NS398). baicalein 190-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-211 15922018-2 2005 FMO, like cytochrome P450 (CYP), is a monooxygenase, utilizing the reducing equivalents of NADPH to reduce 1 atom of molecular oxygen to water, while the other atom is used to oxidize the substrate. NADP 91-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 15922018-2 2005 FMO, like cytochrome P450 (CYP), is a monooxygenase, utilizing the reducing equivalents of NADPH to reduce 1 atom of molecular oxygen to water, while the other atom is used to oxidize the substrate. NADP 91-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 15854816-1 2005 Cytochrome P450 (CYP) is a large family of enzymes containing heme as the active site. Heme 62-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15854816-1 2005 Cytochrome P450 (CYP) is a large family of enzymes containing heme as the active site. Heme 62-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 15769884-1 2005 The polymorphic human cytochrome P450 (P450) 2B6 is primarily responsible for the metabolism of several clinically relevant drugs including bupropion, cyclophosphamide, propofol, and efavirenz. Bupropion 140-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-48 15769884-1 2005 The polymorphic human cytochrome P450 (P450) 2B6 is primarily responsible for the metabolism of several clinically relevant drugs including bupropion, cyclophosphamide, propofol, and efavirenz. Cyclophosphamide 151-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-48 15769884-1 2005 The polymorphic human cytochrome P450 (P450) 2B6 is primarily responsible for the metabolism of several clinically relevant drugs including bupropion, cyclophosphamide, propofol, and efavirenz. Propofol 169-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-48 15769884-1 2005 The polymorphic human cytochrome P450 (P450) 2B6 is primarily responsible for the metabolism of several clinically relevant drugs including bupropion, cyclophosphamide, propofol, and efavirenz. efavirenz 183-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-48 15922018-2 2005 FMO, like cytochrome P450 (CYP), is a monooxygenase, utilizing the reducing equivalents of NADPH to reduce 1 atom of molecular oxygen to water, while the other atom is used to oxidize the substrate. Oxygen 42-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 15922018-2 2005 FMO, like cytochrome P450 (CYP), is a monooxygenase, utilizing the reducing equivalents of NADPH to reduce 1 atom of molecular oxygen to water, while the other atom is used to oxidize the substrate. Oxygen 42-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 15922018-2 2005 FMO, like cytochrome P450 (CYP), is a monooxygenase, utilizing the reducing equivalents of NADPH to reduce 1 atom of molecular oxygen to water, while the other atom is used to oxidize the substrate. Water 137-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 15922018-2 2005 FMO, like cytochrome P450 (CYP), is a monooxygenase, utilizing the reducing equivalents of NADPH to reduce 1 atom of molecular oxygen to water, while the other atom is used to oxidize the substrate. Water 137-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 15840428-0 2005 Lack of evidence for metabolism of p-phenylenediamine by human hepatic cytochrome P450 enzymes. 4-phenylenediamine 35-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 15840428-1 2005 p-Phenylenediamine (PPD) is a widely used ingredient in permanent hair dyes; however, little has been published on its metabolism, especially with respect to hepatic cytochrome P450 (CYP)-mediated oxidation. 4-phenylenediamine 0-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-181 15840428-1 2005 p-Phenylenediamine (PPD) is a widely used ingredient in permanent hair dyes; however, little has been published on its metabolism, especially with respect to hepatic cytochrome P450 (CYP)-mediated oxidation. 4-phenylenediamine 0-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-186 15840428-1 2005 p-Phenylenediamine (PPD) is a widely used ingredient in permanent hair dyes; however, little has been published on its metabolism, especially with respect to hepatic cytochrome P450 (CYP)-mediated oxidation. 4-phenylenediamine 20-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-181 15840428-1 2005 p-Phenylenediamine (PPD) is a widely used ingredient in permanent hair dyes; however, little has been published on its metabolism, especially with respect to hepatic cytochrome P450 (CYP)-mediated oxidation. 4-phenylenediamine 20-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-186 15840428-6 2005 In contrast, 2-aminofluorene underwent CYP-mediated metabolism to > or = 4 different hydroxylated metabolites. 2-aminofluorene 13-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 15772082-1 2005 Testosterone 6beta-hydroxylation is a prototypic reaction of cytochrome P450 (P450) 3A4, the major human P450. testosterone 6beta 0-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-87 16192110-0 2005 Identification of cytochrome P450 forms involved in the 4-hydroxylation of valsartan, a potent and specific angiotensin II receptor antagonist, in human liver microsomes. Valsartan 75-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 16192110-3 2005 The current in vitro studies were conducted to identify the cytochrome P450 (CYP) enzymes involved in the formation of 4-OH valsartan. 4-oh valsartan 119-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 16192110-3 2005 The current in vitro studies were conducted to identify the cytochrome P450 (CYP) enzymes involved in the formation of 4-OH valsartan. 4-oh valsartan 119-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-80 16192110-9 2005 For the specific CYP inhibitors or substrates examined (furafylline, diclofenac, S(+)-mephenytoin, quinidine and troleandomycin), only diclofenac inhibited the formation of 4-OH valsartan. furafylline 56-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 16192110-9 2005 For the specific CYP inhibitors or substrates examined (furafylline, diclofenac, S(+)-mephenytoin, quinidine and troleandomycin), only diclofenac inhibited the formation of 4-OH valsartan. Troleandomycin 113-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 16192110-9 2005 For the specific CYP inhibitors or substrates examined (furafylline, diclofenac, S(+)-mephenytoin, quinidine and troleandomycin), only diclofenac inhibited the formation of 4-OH valsartan. Diclofenac 135-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 16192110-9 2005 For the specific CYP inhibitors or substrates examined (furafylline, diclofenac, S(+)-mephenytoin, quinidine and troleandomycin), only diclofenac inhibited the formation of 4-OH valsartan. 4-oh valsartan 173-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 16192110-11 2005 Although CYP2C9 is involved in valsartan metabolism, CYP-mediated drug-drug interaction between valsartan and other co-administered drugs would be negligible. Valsartan 31-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 15836631-2 2005 CYP1A1, a cytochrome P-450 enzyme, bioactivates polycyclic aromatic hydrocarbons to reactive metabolite(s) that bind to DNA and initiate carcinogenesis. Polycyclic Aromatic Hydrocarbons 48-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 15986265-5 2005 Before subjecting patients on methadone to other drugs, the QT-interval should be determined and it should be ascertained whether the new agent has the property to prolong the QT-interval or is metabolised by the cytochrome-P450 system. Methadone 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 213-228 15842554-1 2005 AIMS: We evaluated the involvement of cytochrome P450 (CYP) isoforms 2C9 and 2C19 in chlorpropamide 2-hydroxylation in vitro and in chlorpropamide disposition in vivo. Chlorpropamide 85-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 15842554-1 2005 AIMS: We evaluated the involvement of cytochrome P450 (CYP) isoforms 2C9 and 2C19 in chlorpropamide 2-hydroxylation in vitro and in chlorpropamide disposition in vivo. Chlorpropamide 85-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 15842554-2 2005 METHODS: To identify CYP isoforms(s) that catalyse 2-hydroxylation of chlorpropamide, the incubation studies were conducted using human liver microsomes and recombinant CYP isoforms. Chlorpropamide 70-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 15842554-2 2005 METHODS: To identify CYP isoforms(s) that catalyse 2-hydroxylation of chlorpropamide, the incubation studies were conducted using human liver microsomes and recombinant CYP isoforms. Chlorpropamide 70-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 15842554-5 2005 In incubation studies using human recombinant CYP isoforms, however, 2-OH-chlorpropamide was formed by both CYP2C9 and CYP2C19 with similar intrinsic clearances (CYP2C9 vs. CYP2C19: 0.26 vs. 0.22 microl min(-1) nmol(-1) protein). CHEBI:143296 69-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 15778010-0 2005 Inhibition of human cytochrome P450 enzymes by the natural hepatotoxin safrole. Safrole 71-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 15863898-0 2005 Inhibitory effects of nicardipine to cytochrome P450 (CYP) in human liver microsomes. Nicardipine 22-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 15863898-0 2005 Inhibitory effects of nicardipine to cytochrome P450 (CYP) in human liver microsomes. Nicardipine 22-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 15863898-1 2005 To anticipate drug-drug interactions by nicardipine in vivo, cytochrome P450 (CYP) forms responsible for the metabolism of nicardipine and inhibition of CYP-dependent drug metabolism by nicardipine were investigated. Nicardipine 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 15863898-1 2005 To anticipate drug-drug interactions by nicardipine in vivo, cytochrome P450 (CYP) forms responsible for the metabolism of nicardipine and inhibition of CYP-dependent drug metabolism by nicardipine were investigated. Nicardipine 123-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 15863898-1 2005 To anticipate drug-drug interactions by nicardipine in vivo, cytochrome P450 (CYP) forms responsible for the metabolism of nicardipine and inhibition of CYP-dependent drug metabolism by nicardipine were investigated. Nicardipine 123-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 15863898-1 2005 To anticipate drug-drug interactions by nicardipine in vivo, cytochrome P450 (CYP) forms responsible for the metabolism of nicardipine and inhibition of CYP-dependent drug metabolism by nicardipine were investigated. Nicardipine 123-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 15863898-1 2005 To anticipate drug-drug interactions by nicardipine in vivo, cytochrome P450 (CYP) forms responsible for the metabolism of nicardipine and inhibition of CYP-dependent drug metabolism by nicardipine were investigated. Nicardipine 123-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 15863898-2 2005 Microsomes of human B-lymphoblastoid cells expressing each human CYP form were used for the metabolism of nicardipine. Nicardipine 106-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 15863898-4 2005 CYP2C8, CYP2D6 and CYP3A4 were identified as major CYP forms for the metabolism of nicardipine in human liver microsomes. Nicardipine 83-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 15769614-1 2005 In breast cancer, cytochrome P450 (CYP) metabolizes both endogenous substrates (i.e. estradiol) and exogenous substrates (i.e. anticancer drugs), which is associated not only with tumor development and progression but also with efficacy of cancer treatment. Estradiol 85-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 15769614-1 2005 In breast cancer, cytochrome P450 (CYP) metabolizes both endogenous substrates (i.e. estradiol) and exogenous substrates (i.e. anticancer drugs), which is associated not only with tumor development and progression but also with efficacy of cancer treatment. Estradiol 85-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 16012077-0 2005 Identification of human cytochrome P450 enzymes involved in the formation of 4-hydroxyestazolam from estazolam. 4-Hydroxy Estazolam 77-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 15843388-0 2005 Escherichia coli BTC, a human cytochrome P450 competent tester strain with a high sensitivity towards alkylating agents: involvement of alkyltransferases in the repair of DNA damage induced by aromatic amines. aromatic amines 193-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 16012077-0 2005 Identification of human cytochrome P450 enzymes involved in the formation of 4-hydroxyestazolam from estazolam. Estazolam 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 16012077-1 2005 To predict drug interactions with estazolam, the biotransformation of estazolam to its major hydoxylated metabolite, 4-hydroxyestazolam was studied in vitro using pooled human liver microsomes and individual expressed human cytochrome P450 (CYP) enzymes. Estazolam 70-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-244 16012077-5 2005 When estazolam was incubated with expressed human CYP enzymes (CYP1A2, CYP2A6, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4), it was metabolized only by CYP3A4. Estazolam 5-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 16833677-5 2005 This thiolate complex is used as a mimic for the cytochrome P450s active site to model the first step of the catalytic cycle of this enzyme. thiolate 5-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 16833677-8 2005 These calculations are in line with the experimentally observed change in the spin state from low to high spin upon this removal of the axial hydroxo ligand by binding of the substrate in the heme pocket of cytochrome P450. Heme 192-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-222 15876406-0 2005 New assumptions about oxidative processes involved in steroid hormone biosynthesis: is the role of cytochrome P-450-activated dioxygen limited to hydroxylation reactions or are dioxygen insertion reactions also possible? Steroids 54-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 15576430-8 2005 Our results suggest that the cerebral endothelial barrier dysfunction seen in response to glutamate is Ca(2+) dependent and may require several intracellular signaling events mediated by oxidants derived from reduced nicotinamide adenine dinucleotide oxidase, cytochrome P-450, and the mitochondria. Glutamic Acid 90-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 260-276 15665333-1 2005 Human cytochrome P450 (P450) 2A6 catalyzes 7-hydroxylation of coumarin, and the reaction rate is enhanced by cytochrome b5 (b5). coumarin 62-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-32 15876406-0 2005 New assumptions about oxidative processes involved in steroid hormone biosynthesis: is the role of cytochrome P-450-activated dioxygen limited to hydroxylation reactions or are dioxygen insertion reactions also possible? Oxygen 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 15784278-0 2005 NADPH-dependent metabolism of 17beta-estradiol and estrone to polar and nonpolar metabolites by human tissues and cytochrome P450 isoforms. Estradiol 30-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 15772515-8 2005 These findings suggest that nicorandil causes vasodilation in forearm circulation in humans, at least in part through a pathway that is dependent on K-ATP channels and cytochrome P-450, but not on endogenous NO and prostaglandins. Nicorandil 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-184 15784278-3 2005 Precise knowledge of the CYP-mediated formation of these metabolites, particularly those with unique biological activities (e.g., 4-hydroxy-E(2), 16alpha-hydroxy-E(1), 15alpha-hydroxy-E(2), 16-epiestriol, and 2-methoxyestradiol) in human liver and extrahepatic target tissues and cells, would add significantly to our understanding of the diverse biological functions that are associated with endogenous estrogens. 4-hydroxy-e(2) 130-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 15784278-3 2005 Precise knowledge of the CYP-mediated formation of these metabolites, particularly those with unique biological activities (e.g., 4-hydroxy-E(2), 16alpha-hydroxy-E(1), 15alpha-hydroxy-E(2), 16-epiestriol, and 2-methoxyestradiol) in human liver and extrahepatic target tissues and cells, would add significantly to our understanding of the diverse biological functions that are associated with endogenous estrogens. 16alpha-hydroxy-e( 146-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 15784278-3 2005 Precise knowledge of the CYP-mediated formation of these metabolites, particularly those with unique biological activities (e.g., 4-hydroxy-E(2), 16alpha-hydroxy-E(1), 15alpha-hydroxy-E(2), 16-epiestriol, and 2-methoxyestradiol) in human liver and extrahepatic target tissues and cells, would add significantly to our understanding of the diverse biological functions that are associated with endogenous estrogens. 15alpha-hydroxy-e( 168-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 15784278-3 2005 Precise knowledge of the CYP-mediated formation of these metabolites, particularly those with unique biological activities (e.g., 4-hydroxy-E(2), 16alpha-hydroxy-E(1), 15alpha-hydroxy-E(2), 16-epiestriol, and 2-methoxyestradiol) in human liver and extrahepatic target tissues and cells, would add significantly to our understanding of the diverse biological functions that are associated with endogenous estrogens. ), 16-epiestriol 187-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 15784278-3 2005 Precise knowledge of the CYP-mediated formation of these metabolites, particularly those with unique biological activities (e.g., 4-hydroxy-E(2), 16alpha-hydroxy-E(1), 15alpha-hydroxy-E(2), 16-epiestriol, and 2-methoxyestradiol) in human liver and extrahepatic target tissues and cells, would add significantly to our understanding of the diverse biological functions that are associated with endogenous estrogens. 2-Methoxyestradiol 209-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 15784278-4 2005 In this article, we review recent results on the NADPH-dependent metabolism of endogenous estrogens to polar (hydroxylated and keto) metabolites as well as to nonpolar metabolites by human tissues and recombinant human CYP isoforms. NADP 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 219-222 15784278-5 2005 The available data show that a large number of polar and nonpolar metabolites of E(2) and E(1) are formed by human tissues, and a variety of human CYP isoforms are involved in the NADPH-dependent formation of polar as well as nonpolar estrogen metabolites. NADP 180-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 15713371-0 2005 Effects of suberoylanilide hydroxamic acid and trichostatin A on induction of cytochrome P450 enzymes and benzo[a]pyrene DNA adduct formation in human cells. Vorinostat 11-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 15750390-2 2005 This research examined the impact of single nucleotide polymorphisms (SNP) of MDR1 and CYP genes on nelfinavir and efavirenz pharmacokinetics and the response to highly active antiretroviral therapy (HAART) in HIV-1 infected children. Nelfinavir 100-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 15713371-0 2005 Effects of suberoylanilide hydroxamic acid and trichostatin A on induction of cytochrome P450 enzymes and benzo[a]pyrene DNA adduct formation in human cells. trichostatin A 47-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 15713371-3 2005 Results from our study indicated a significant increase in CYP activity in comparison to vehicle control in cells treated with SAHA or TSA as measured by ethoxyresorufin-O-deethylase assay. trichostatin A 135-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 15608133-0 2005 Cytochrome P450-mediated oxidation of glucuronide derivatives: example of estradiol-17beta-glucuronide oxidation to 2-hydroxy-estradiol-17beta-glucuronide by CYP 2C8. Glucuronides 38-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15713371-8 2005 These results suggest that while CYP enzyme activity and gene expression were affected by the HDAC inhibitors SAHA and TSA, they had no apparent influence on B[a]P DNA binding. trichostatin A 119-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 15608133-2 2005 Cytochrome P450-mediated oxidation of steroid sulfate has been described, suggesting that oxidation of polar metabolites such as glucuronide derivatives of endogenous compounds can occur. steroid sulfate 38-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15608133-2 2005 Cytochrome P450-mediated oxidation of steroid sulfate has been described, suggesting that oxidation of polar metabolites such as glucuronide derivatives of endogenous compounds can occur. Glucuronides 129-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15608133-7 2005 This is the first demonstration of cytochrome P450-mediated oxidation of a steroid glucuro-conjugate. Steroids 75-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 15616733-0 2005 Biomimetic alcohol oxidations by an iron(III) porphyrin complex: relevance to cytochrome P-450 catalytic oxidation and involvement of the two-state radical rebound mechanism. Alcohols 11-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 16019948-0 2005 Identification of cytochrome P450 enzymes involved in the metabolism of 3",4"-methylenedioxy-alpha-pyrrolidinopropiophenone (MDPPP), a designer drug, in human liver microsomes. 3',4'-methylenedioxy-alpha-pyrrolidinopropiophenone 72-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 16019948-0 2005 Identification of cytochrome P450 enzymes involved in the metabolism of 3",4"-methylenedioxy-alpha-pyrrolidinopropiophenone (MDPPP), a designer drug, in human liver microsomes. 3',4'-methylenedioxy-alpha-pyrrolidinopropiophenone 125-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 16019948-1 2005 The metabolism of 3",4"-methylenedioxy-a-pyrrolidinopropiophenone (MDPPP), a novel designer drug, to its demethylenated major metabolite 3",4"-dihydroxy-pyrrolidinopropiophenone (di-HO-PPP) was studied in pooled human liver microsomes (HLM) and in cDNA-expressed human hepatic cytochrome P450 (CYP) enzymes. 3",4"-methylenedioxy-a-pyrrolidinopropiophenone 18-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 277-292 16019948-1 2005 The metabolism of 3",4"-methylenedioxy-a-pyrrolidinopropiophenone (MDPPP), a novel designer drug, to its demethylenated major metabolite 3",4"-dihydroxy-pyrrolidinopropiophenone (di-HO-PPP) was studied in pooled human liver microsomes (HLM) and in cDNA-expressed human hepatic cytochrome P450 (CYP) enzymes. 3",4"-methylenedioxy-a-pyrrolidinopropiophenone 18-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-297 16019948-1 2005 The metabolism of 3",4"-methylenedioxy-a-pyrrolidinopropiophenone (MDPPP), a novel designer drug, to its demethylenated major metabolite 3",4"-dihydroxy-pyrrolidinopropiophenone (di-HO-PPP) was studied in pooled human liver microsomes (HLM) and in cDNA-expressed human hepatic cytochrome P450 (CYP) enzymes. 3',4'-methylenedioxy-alpha-pyrrolidinopropiophenone 67-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 277-292 16019948-1 2005 The metabolism of 3",4"-methylenedioxy-a-pyrrolidinopropiophenone (MDPPP), a novel designer drug, to its demethylenated major metabolite 3",4"-dihydroxy-pyrrolidinopropiophenone (di-HO-PPP) was studied in pooled human liver microsomes (HLM) and in cDNA-expressed human hepatic cytochrome P450 (CYP) enzymes. 3',4'-methylenedioxy-alpha-pyrrolidinopropiophenone 67-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-297 16019948-1 2005 The metabolism of 3",4"-methylenedioxy-a-pyrrolidinopropiophenone (MDPPP), a novel designer drug, to its demethylenated major metabolite 3",4"-dihydroxy-pyrrolidinopropiophenone (di-HO-PPP) was studied in pooled human liver microsomes (HLM) and in cDNA-expressed human hepatic cytochrome P450 (CYP) enzymes. 3",4"-dihydroxy-pyrrolidinopropiophenone 137-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 277-292 16019948-1 2005 The metabolism of 3",4"-methylenedioxy-a-pyrrolidinopropiophenone (MDPPP), a novel designer drug, to its demethylenated major metabolite 3",4"-dihydroxy-pyrrolidinopropiophenone (di-HO-PPP) was studied in pooled human liver microsomes (HLM) and in cDNA-expressed human hepatic cytochrome P450 (CYP) enzymes. 3",4"-dihydroxy-pyrrolidinopropiophenone 137-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 294-297 15767245-6 2005 We propose that the increased antithrombotic effect of warfarin involved protein-binding interactions and decreased warfarin metabolism through the cytochrome P450 (CYP) enzyme system. Warfarin 55-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-163 15767245-6 2005 We propose that the increased antithrombotic effect of warfarin involved protein-binding interactions and decreased warfarin metabolism through the cytochrome P450 (CYP) enzyme system. Warfarin 55-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-168 15767245-6 2005 We propose that the increased antithrombotic effect of warfarin involved protein-binding interactions and decreased warfarin metabolism through the cytochrome P450 (CYP) enzyme system. Warfarin 116-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-163 15767245-6 2005 We propose that the increased antithrombotic effect of warfarin involved protein-binding interactions and decreased warfarin metabolism through the cytochrome P450 (CYP) enzyme system. Warfarin 116-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-168 15767245-7 2005 Concurrent administration of aspirin and other drugs that are metabolized through or are inhibitors of the CYP system may have enhanced the interaction that occurred in this patient. Aspirin 29-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-110 15746054-2 2005 Cytochrome P450 enzymes are primarily expressed in the liver and convert the prodrug cyclophosphamide to an active phosphoramide mustard and acrolein. Cyclophosphamide 85-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15746054-2 2005 Cytochrome P450 enzymes are primarily expressed in the liver and convert the prodrug cyclophosphamide to an active phosphoramide mustard and acrolein. phosphoramide 115-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15746054-2 2005 Cytochrome P450 enzymes are primarily expressed in the liver and convert the prodrug cyclophosphamide to an active phosphoramide mustard and acrolein. Acrolein 141-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15596250-1 2005 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is highly toxic in experimental animals, and is known to induce cytochrome P450 (CYP) gene expression. Polychlorinated Dibenzodioxins 0-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 15596250-1 2005 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is highly toxic in experimental animals, and is known to induce cytochrome P450 (CYP) gene expression. Polychlorinated Dibenzodioxins 0-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 15596250-1 2005 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is highly toxic in experimental animals, and is known to induce cytochrome P450 (CYP) gene expression. Polychlorinated Dibenzodioxins 37-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 15596250-1 2005 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is highly toxic in experimental animals, and is known to induce cytochrome P450 (CYP) gene expression. Polychlorinated Dibenzodioxins 37-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 15596250-7 2005 However, when lymphocytes were treated in vitro with 10 nM TCDD, CYP1B1 and CYP1A1 mRNA expression was strongly induced and modified by CYP variant alleles. Polychlorinated Dibenzodioxins 59-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 15596250-11 2005 Genetic variation in cytochrome P450 induction may identify subjects with variable responsiveness to TCDD and potentially increased risk of disease. Polychlorinated Dibenzodioxins 101-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 15616733-0 2005 Biomimetic alcohol oxidations by an iron(III) porphyrin complex: relevance to cytochrome P-450 catalytic oxidation and involvement of the two-state radical rebound mechanism. iron(iii) porphyrin 36-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 15616733-1 2005 The systematic oxidation reactions of a wide range of alcohols have been carried out by using an iron porphyrin complex in order to understand their relation to cytochrome P-450 enzymes and to have a practical application to organic synthesis. Alcohols 54-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 15616733-1 2005 The systematic oxidation reactions of a wide range of alcohols have been carried out by using an iron porphyrin complex in order to understand their relation to cytochrome P-450 enzymes and to have a practical application to organic synthesis. iron porphyrin 97-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 15596297-2 2005 TAM is metabolized by cytochrome P450 (CYP450) enzymes, including CYP3A5. Tamoxifen 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 15581596-2 2005 Following intracellular metabolism by cytochrome P450 (CYP) enzymes, drug metabolites are excreted into bile or urine via ATP-dependent multidrug resistance proteins (MDR1 and MRPs). Adenosine Triphosphate 122-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 15581596-2 2005 Following intracellular metabolism by cytochrome P450 (CYP) enzymes, drug metabolites are excreted into bile or urine via ATP-dependent multidrug resistance proteins (MDR1 and MRPs). Adenosine Triphosphate 122-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 15585365-4 2005 The specific CYP isozymes responsible for KR-60436 oxidation to four major metabolites, pyrrole-KR-60436, O-demethylpyrrole-KR-60436, N-dehydroxyethyl-KR-60436 and an active metabolite, O-demethyl-KR-60436 were identified using the combination of correlation analysis, immuno-inhibition, chemical inhibition in human liver microsomes and metabolism by expressed recombinant CYP enzymes. Krypton 42-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 15585365-4 2005 The specific CYP isozymes responsible for KR-60436 oxidation to four major metabolites, pyrrole-KR-60436, O-demethylpyrrole-KR-60436, N-dehydroxyethyl-KR-60436 and an active metabolite, O-demethyl-KR-60436 were identified using the combination of correlation analysis, immuno-inhibition, chemical inhibition in human liver microsomes and metabolism by expressed recombinant CYP enzymes. Krypton 42-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 374-377 15585365-4 2005 The specific CYP isozymes responsible for KR-60436 oxidation to four major metabolites, pyrrole-KR-60436, O-demethylpyrrole-KR-60436, N-dehydroxyethyl-KR-60436 and an active metabolite, O-demethyl-KR-60436 were identified using the combination of correlation analysis, immuno-inhibition, chemical inhibition in human liver microsomes and metabolism by expressed recombinant CYP enzymes. pyrrole-kr 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 15585365-4 2005 The specific CYP isozymes responsible for KR-60436 oxidation to four major metabolites, pyrrole-KR-60436, O-demethylpyrrole-KR-60436, N-dehydroxyethyl-KR-60436 and an active metabolite, O-demethyl-KR-60436 were identified using the combination of correlation analysis, immuno-inhibition, chemical inhibition in human liver microsomes and metabolism by expressed recombinant CYP enzymes. o-demethylpyrrole-kr 106-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 15585365-4 2005 The specific CYP isozymes responsible for KR-60436 oxidation to four major metabolites, pyrrole-KR-60436, O-demethylpyrrole-KR-60436, N-dehydroxyethyl-KR-60436 and an active metabolite, O-demethyl-KR-60436 were identified using the combination of correlation analysis, immuno-inhibition, chemical inhibition in human liver microsomes and metabolism by expressed recombinant CYP enzymes. n-dehydroxyethyl-kr 134-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 15585365-4 2005 The specific CYP isozymes responsible for KR-60436 oxidation to four major metabolites, pyrrole-KR-60436, O-demethylpyrrole-KR-60436, N-dehydroxyethyl-KR-60436 and an active metabolite, O-demethyl-KR-60436 were identified using the combination of correlation analysis, immuno-inhibition, chemical inhibition in human liver microsomes and metabolism by expressed recombinant CYP enzymes. o-demethyl-kr 186-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 15596297-2 2005 TAM is metabolized by cytochrome P450 (CYP450) enzymes, including CYP3A5. Tamoxifen 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-45 16175235-2 2005 Ifosfamide must be activated by cytochrome P450 (P450) enzymes in the liver, initially to a short lived intermediate and then to toxic metabolites that are subsequently distributed by the circulatory system. Ifosfamide 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 15630035-0 2005 Cytochrome P-450 under pressure: more evidence for a link between 20-hydroxyeicosatetraenoic acid and hypertension. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 66-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 15740177-4 2005 Furthermore, although dolasetron, granisetron, ondansetron and tropisetron are all extensively metabolised by the cytochrome P450 (CYP) system, different components of this system predominate in the metabolism of each of these agents. dolasetron 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 15635191-1 2005 The effects of sulpiride, an antipsychotic drug, on cytochrome P450 (CYP) activities in human liver microsomes were investigated. Sulpiride 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 15635191-1 2005 The effects of sulpiride, an antipsychotic drug, on cytochrome P450 (CYP) activities in human liver microsomes were investigated. Sulpiride 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 15606443-1 2005 AIMS: Rosiglitazone, a thiazolidinedione antidiabetic medication used in the treatment of Type 2 diabetes mellitus, is predominantly metabolized by the cytochrome P450 (CYP) enzyme CYP2C8. Rosiglitazone 6-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-167 15606443-1 2005 AIMS: Rosiglitazone, a thiazolidinedione antidiabetic medication used in the treatment of Type 2 diabetes mellitus, is predominantly metabolized by the cytochrome P450 (CYP) enzyme CYP2C8. Rosiglitazone 6-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 15606443-1 2005 AIMS: Rosiglitazone, a thiazolidinedione antidiabetic medication used in the treatment of Type 2 diabetes mellitus, is predominantly metabolized by the cytochrome P450 (CYP) enzyme CYP2C8. 2,4-thiazolidinedione 23-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-167 15606443-1 2005 AIMS: Rosiglitazone, a thiazolidinedione antidiabetic medication used in the treatment of Type 2 diabetes mellitus, is predominantly metabolized by the cytochrome P450 (CYP) enzyme CYP2C8. 2,4-thiazolidinedione 23-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 15635179-0 2005 Soybean oil fat emulsion prevents cytochrome P450 mRNA down-regulation induced by fat-free overdose total parenteral nutrition in infant rats. Oils 8-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 15665498-0 2005 Interaction of modified cyclodextrins with cytochrome P-450. Cyclodextrins 24-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 15665498-1 2005 The effects of modified cyclodextrins (CDs) hydroxypropyl-beta-CD and methyl-beta-CD were studied in vitro on cDNA-expressed human cytochrome P-450 (CYP) activities (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). Cyclodextrins 24-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 15665498-1 2005 The effects of modified cyclodextrins (CDs) hydroxypropyl-beta-CD and methyl-beta-CD were studied in vitro on cDNA-expressed human cytochrome P-450 (CYP) activities (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). Cyclodextrins 24-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-152 15665498-1 2005 The effects of modified cyclodextrins (CDs) hydroxypropyl-beta-CD and methyl-beta-CD were studied in vitro on cDNA-expressed human cytochrome P-450 (CYP) activities (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). Cyclodextrins 39-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 15665498-1 2005 The effects of modified cyclodextrins (CDs) hydroxypropyl-beta-CD and methyl-beta-CD were studied in vitro on cDNA-expressed human cytochrome P-450 (CYP) activities (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). methyl-beta-cyclodextrin 70-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 16176119-2 2005 Preclinical studies were conducted to determine the cytochrome P450 (CYP) isoenzymes involved in the metabolism of gefitinib and to evaluate the potential of gefitinib to cause drug interactions through inhibition of CYP isoenzymes. Gefitinib 115-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 16176119-2 2005 Preclinical studies were conducted to determine the cytochrome P450 (CYP) isoenzymes involved in the metabolism of gefitinib and to evaluate the potential of gefitinib to cause drug interactions through inhibition of CYP isoenzymes. Gefitinib 115-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 16176119-2 2005 Preclinical studies were conducted to determine the cytochrome P450 (CYP) isoenzymes involved in the metabolism of gefitinib and to evaluate the potential of gefitinib to cause drug interactions through inhibition of CYP isoenzymes. Gefitinib 158-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 217-220 16176119-4 2005 METHODS: In preclinical studies radiolabelled gefitinib was incubated with: (i) hepatic microsomal protein in the presence of selective CYP inhibitors; and (ii) expressed CYP enzymes. Gefitinib 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-139 16176119-4 2005 METHODS: In preclinical studies radiolabelled gefitinib was incubated with: (i) hepatic microsomal protein in the presence of selective CYP inhibitors; and (ii) expressed CYP enzymes. Gefitinib 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-174 16176119-5 2005 Human hepatic microsomal protein was incubated with selective CYP substrates in the presence of gefitinib. Gefitinib 96-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 15740177-4 2005 Furthermore, although dolasetron, granisetron, ondansetron and tropisetron are all extensively metabolised by the cytochrome P450 (CYP) system, different components of this system predominate in the metabolism of each of these agents. dolasetron 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 15740177-4 2005 Furthermore, although dolasetron, granisetron, ondansetron and tropisetron are all extensively metabolised by the cytochrome P450 (CYP) system, different components of this system predominate in the metabolism of each of these agents. Granisetron 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 15740177-4 2005 Furthermore, although dolasetron, granisetron, ondansetron and tropisetron are all extensively metabolised by the cytochrome P450 (CYP) system, different components of this system predominate in the metabolism of each of these agents. Granisetron 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 15740177-4 2005 Furthermore, although dolasetron, granisetron, ondansetron and tropisetron are all extensively metabolised by the cytochrome P450 (CYP) system, different components of this system predominate in the metabolism of each of these agents. Ondansetron 47-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 15740177-4 2005 Furthermore, although dolasetron, granisetron, ondansetron and tropisetron are all extensively metabolised by the cytochrome P450 (CYP) system, different components of this system predominate in the metabolism of each of these agents. Ondansetron 47-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 15638760-5 2005 In other contemporary studies, screening hypericin for inhibitory effects on various pharmaceutically important enzymes such as MAO (monoaminoxidase), PKC (protein kinase C), dopamine-beta-hydroxylase, reverse transcriptase, telomerase and CYP (cytochrome P450), has yielded results supporting therapeutic potential. hypericin 41-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-243 15638760-5 2005 In other contemporary studies, screening hypericin for inhibitory effects on various pharmaceutically important enzymes such as MAO (monoaminoxidase), PKC (protein kinase C), dopamine-beta-hydroxylase, reverse transcriptase, telomerase and CYP (cytochrome P450), has yielded results supporting therapeutic potential. hypericin 41-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 245-260 15740177-4 2005 Furthermore, although dolasetron, granisetron, ondansetron and tropisetron are all extensively metabolised by the cytochrome P450 (CYP) system, different components of this system predominate in the metabolism of each of these agents. Tropisetron 63-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 15740177-4 2005 Furthermore, although dolasetron, granisetron, ondansetron and tropisetron are all extensively metabolised by the cytochrome P450 (CYP) system, different components of this system predominate in the metabolism of each of these agents. Tropisetron 63-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 15963007-13 2005 In contrast, sulphonylureas, meglitinide derivatives and thiazolidinediones are extensively metabolised in the liver via the CYP system and thus, may be subject to drug-drug metabolic interactions. meglitinide 29-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 15963007-13 2005 In contrast, sulphonylureas, meglitinide derivatives and thiazolidinediones are extensively metabolised in the liver via the CYP system and thus, may be subject to drug-drug metabolic interactions. Thiazolidinediones 57-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 15963007-20 2005 The effects of inducers or inhibitors of CYP isoenzymes on the metabolism and pharmacokinetics of the glucose-lowering agents of each pharmacological class has been tested. Glucose 102-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 15963007-21 2005 Significantly increased (with CYP inhibitors) or decreased (with CYP inducers) plasma levels of sulphonylureas, meglitinide derivatives and thiazolidinediones have been reported in healthy volunteers, and these pharmacokinetic changes may lead to enhanced or reduced glucose-lowering action, and thus hypoglycaemia or worsening of metabolic control, respectively. meglitinide 112-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 16393888-6 2005 Both CPA and IFO are prodrugs that require activation by hepatic cytochrome P450 (CYP)-catalyzed 4-hydroxylation, yielding cytotoxic nitrogen mustards capable of reacting with DNA molecules to form crosslinks and lead to cell apoptosis and/or necrosis. Cyclophosphamide 5-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 15963007-21 2005 Significantly increased (with CYP inhibitors) or decreased (with CYP inducers) plasma levels of sulphonylureas, meglitinide derivatives and thiazolidinediones have been reported in healthy volunteers, and these pharmacokinetic changes may lead to enhanced or reduced glucose-lowering action, and thus hypoglycaemia or worsening of metabolic control, respectively. Thiazolidinediones 140-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 16393888-6 2005 Both CPA and IFO are prodrugs that require activation by hepatic cytochrome P450 (CYP)-catalyzed 4-hydroxylation, yielding cytotoxic nitrogen mustards capable of reacting with DNA molecules to form crosslinks and lead to cell apoptosis and/or necrosis. Cyclophosphamide 5-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 15895287-1 2005 The cytochrome P450 (CYP) is a monooxidase, which regulates metabolism of drugs and fatty acids in the liver and kidney. Fatty Acids 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 16393888-6 2005 Both CPA and IFO are prodrugs that require activation by hepatic cytochrome P450 (CYP)-catalyzed 4-hydroxylation, yielding cytotoxic nitrogen mustards capable of reacting with DNA molecules to form crosslinks and lead to cell apoptosis and/or necrosis. Nitrogen 133-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 16393888-6 2005 Both CPA and IFO are prodrugs that require activation by hepatic cytochrome P450 (CYP)-catalyzed 4-hydroxylation, yielding cytotoxic nitrogen mustards capable of reacting with DNA molecules to form crosslinks and lead to cell apoptosis and/or necrosis. Nitrogen 133-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 16393888-11 2005 The alternative CYP-catalyzed inactivation pathway by N-dechloroethylation generates the neurotoxic and nephrotoxic byproduct chloroacetaldehyde (CAA). Nitrogen 54-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 16393888-11 2005 The alternative CYP-catalyzed inactivation pathway by N-dechloroethylation generates the neurotoxic and nephrotoxic byproduct chloroacetaldehyde (CAA). chloroacetaldehyde 126-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 16393888-11 2005 The alternative CYP-catalyzed inactivation pathway by N-dechloroethylation generates the neurotoxic and nephrotoxic byproduct chloroacetaldehyde (CAA). chloroacetaldehyde 146-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 21783461-0 2005 Formation of estrogenic metabolites of benzo[a]pyrene and chrysene by cytochrome P450 activity and their combined and supra-maximal estrogenic activity. Benzo(a)pyrene 39-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 21783461-0 2005 Formation of estrogenic metabolites of benzo[a]pyrene and chrysene by cytochrome P450 activity and their combined and supra-maximal estrogenic activity. chrysene 58-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 21783461-8 2005 BaP itself exhibited estrogenicity in the ER-CALUX assay due to bio-activation into estrogenic metabolites, probably via aryl hydrocarbon receptor (AhR) induced CYP activity. Benzo(a)pyrene 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 16363885-13 2005 Biotransformation by the cytochrome P450 (CYP450) system is an important step in the activation or elimination of a large number of drugs, including oxybutynin, tolterodine, darifenacin and solifenacin, raising the possibility of clinically relevant and potentially serious drug interactions. oxybutynin 149-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 16363885-13 2005 Biotransformation by the cytochrome P450 (CYP450) system is an important step in the activation or elimination of a large number of drugs, including oxybutynin, tolterodine, darifenacin and solifenacin, raising the possibility of clinically relevant and potentially serious drug interactions. oxybutynin 149-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-48 16363885-13 2005 Biotransformation by the cytochrome P450 (CYP450) system is an important step in the activation or elimination of a large number of drugs, including oxybutynin, tolterodine, darifenacin and solifenacin, raising the possibility of clinically relevant and potentially serious drug interactions. Tolterodine Tartrate 161-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 16363885-13 2005 Biotransformation by the cytochrome P450 (CYP450) system is an important step in the activation or elimination of a large number of drugs, including oxybutynin, tolterodine, darifenacin and solifenacin, raising the possibility of clinically relevant and potentially serious drug interactions. Tolterodine Tartrate 161-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-48 16363885-13 2005 Biotransformation by the cytochrome P450 (CYP450) system is an important step in the activation or elimination of a large number of drugs, including oxybutynin, tolterodine, darifenacin and solifenacin, raising the possibility of clinically relevant and potentially serious drug interactions. darifenacin 174-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 16363885-13 2005 Biotransformation by the cytochrome P450 (CYP450) system is an important step in the activation or elimination of a large number of drugs, including oxybutynin, tolterodine, darifenacin and solifenacin, raising the possibility of clinically relevant and potentially serious drug interactions. darifenacin 174-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-48 16363885-13 2005 Biotransformation by the cytochrome P450 (CYP450) system is an important step in the activation or elimination of a large number of drugs, including oxybutynin, tolterodine, darifenacin and solifenacin, raising the possibility of clinically relevant and potentially serious drug interactions. Solifenacin Succinate 190-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 16363885-13 2005 Biotransformation by the cytochrome P450 (CYP450) system is an important step in the activation or elimination of a large number of drugs, including oxybutynin, tolterodine, darifenacin and solifenacin, raising the possibility of clinically relevant and potentially serious drug interactions. Solifenacin Succinate 190-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-48 16329398-0 2005 [Ratio of low- and high-spin cytochrome P-450 in liver microsomes of N-nitrosodiethylamine-induced hepatomas]. Diethylnitrosamine 69-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 15895287-1 2005 The cytochrome P450 (CYP) is a monooxidase, which regulates metabolism of drugs and fatty acids in the liver and kidney. Fatty Acids 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 16116487-1 2005 Cyclophosphamide (CP), a widely used cytostatic, is metabolized by polymorphic drug metabolizing enzymes particularly cytochrome P450 (CYP) enzymes. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 16265100-4 2005 An increase in oxidative stress, due to the generation of reactive oxygen species as a result of mitochondrial abnormalities and induction of the cytochrome P-450 system could be one mechanism by which the nonalcoholic fatty liver develops into nonalcoholic steatohepatitis. Reactive Oxygen Species 58-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-162 16116487-1 2005 Cyclophosphamide (CP), a widely used cytostatic, is metabolized by polymorphic drug metabolizing enzymes particularly cytochrome P450 (CYP) enzymes. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 15635280-7 2005 In the present review, cytochrome P450 monooxygenases involved in the biosyntheses of three structurally and biosynthetically interesting compounds, secologanin, cornoside, and shikonin, a described together with the results of previous and recent biosynthetic studies. shikonin 177-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 20021073-5 2005 Recent studies have demonstrated that an imbalance between cyclooxygenase-2 (COX-2) and cytochrome P450 (CYP450) arachidonic acid metabolizing enzymes in the kidney may contribute to the renal damage associated with obesity. Arachidonic Acid 113-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 20021073-5 2005 Recent studies have demonstrated that an imbalance between cyclooxygenase-2 (COX-2) and cytochrome P450 (CYP450) arachidonic acid metabolizing enzymes in the kidney may contribute to the renal damage associated with obesity. Arachidonic Acid 113-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-111 15560889-6 2004 The affinity of carbofuran for CYP enzymes involved in the oxidation to 3-hydroxycarbofuran is significantly less in HLM (Km=1.950 mM) than in RLM (Km=0.210 mM), or MLM (Km=0.550 mM). Carbofuran 16-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 15593150-0 2004 An oxidative phenol coupling reaction catalyzed by oxyB, a cytochrome P450 from the vancomycin-producing microorganism. Vancomycin 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 15635280-7 2005 In the present review, cytochrome P450 monooxygenases involved in the biosyntheses of three structurally and biosynthetically interesting compounds, secologanin, cornoside, and shikonin, a described together with the results of previous and recent biosynthetic studies. secologanin 149-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 15635280-7 2005 In the present review, cytochrome P450 monooxygenases involved in the biosyntheses of three structurally and biosynthetically interesting compounds, secologanin, cornoside, and shikonin, a described together with the results of previous and recent biosynthetic studies. cornoside 162-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 15560889-5 2004 Carbofuran is metabolized by cytochrome P450 (CYP) leading to the production of one major ring oxidation metabolite, 3-hydroxycarbofuran, and two minor metabolites. Carbofuran 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 15560889-5 2004 Carbofuran is metabolized by cytochrome P450 (CYP) leading to the production of one major ring oxidation metabolite, 3-hydroxycarbofuran, and two minor metabolites. Carbofuran 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 15560889-6 2004 The affinity of carbofuran for CYP enzymes involved in the oxidation to 3-hydroxycarbofuran is significantly less in HLM (Km=1.950 mM) than in RLM (Km=0.210 mM), or MLM (Km=0.550 mM). 3-hydroxycarbofuran 72-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 15560889-5 2004 Carbofuran is metabolized by cytochrome P450 (CYP) leading to the production of one major ring oxidation metabolite, 3-hydroxycarbofuran, and two minor metabolites. 3-hydroxycarbofuran 117-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 15560889-5 2004 Carbofuran is metabolized by cytochrome P450 (CYP) leading to the production of one major ring oxidation metabolite, 3-hydroxycarbofuran, and two minor metabolites. 3-hydroxycarbofuran 117-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 15560889-8 2004 A screen of 15 major human CYP isoforms for metabolic ability with respect to carbofuran metabolism demonstrated that CYP3A4 is the major isoform responsible for carbofuran oxidation in humans. Carbofuran 78-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 15560889-8 2004 A screen of 15 major human CYP isoforms for metabolic ability with respect to carbofuran metabolism demonstrated that CYP3A4 is the major isoform responsible for carbofuran oxidation in humans. Carbofuran 162-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 15560889-11 2004 Normalization of HLM data with the average levels of each CYP in native HLM, indicates that carbofuran metabolism is primarily mediated by CYP3A4 (percent total normalized rate (% TNR)=77.5), although CYP1A2 and 2C19 play ancillary roles (% TNR=9.0 and 6.0, respectively). Carbofuran 92-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 15560889-15 2004 The variation in carbofuran metabolism among 17 single-donor HLM samples is over 5-fold and the best correlation between CYP isoform activity and carbofuran metabolism was observed with CYP3A4 (r2=0.96). Carbofuran 17-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 15560889-15 2004 The variation in carbofuran metabolism among 17 single-donor HLM samples is over 5-fold and the best correlation between CYP isoform activity and carbofuran metabolism was observed with CYP3A4 (r2=0.96). Carbofuran 146-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 15644946-1 2004 The cytochrome p450 (CYP) superfamily is responsible for the oxidation, peroxidation, and (or) reduction of vitamins, steroids, xenobiotics, and the majority of cardiovascular drugs in an oxygen- and NADPH-dependent manner. NADP 200-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 15519301-1 2004 Alkylresorufins are model substrates for cytochrome P450 (P450) 1A2. alkylresorufins 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-67 15585441-12 2004 As with other protease inhibitors, atazanavir is also a substrate and moderate inhibitor of the cytochrome P450 (CYP) system, in particular CYP3A4 and CYP2C9. Atazanavir Sulfate 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-111 15644946-1 2004 The cytochrome p450 (CYP) superfamily is responsible for the oxidation, peroxidation, and (or) reduction of vitamins, steroids, xenobiotics, and the majority of cardiovascular drugs in an oxygen- and NADPH-dependent manner. Steroids 118-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 15644946-1 2004 The cytochrome p450 (CYP) superfamily is responsible for the oxidation, peroxidation, and (or) reduction of vitamins, steroids, xenobiotics, and the majority of cardiovascular drugs in an oxygen- and NADPH-dependent manner. Steroids 118-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 15644946-1 2004 The cytochrome p450 (CYP) superfamily is responsible for the oxidation, peroxidation, and (or) reduction of vitamins, steroids, xenobiotics, and the majority of cardiovascular drugs in an oxygen- and NADPH-dependent manner. Oxygen 188-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 15644946-1 2004 The cytochrome p450 (CYP) superfamily is responsible for the oxidation, peroxidation, and (or) reduction of vitamins, steroids, xenobiotics, and the majority of cardiovascular drugs in an oxygen- and NADPH-dependent manner. Oxygen 188-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 15644946-1 2004 The cytochrome p450 (CYP) superfamily is responsible for the oxidation, peroxidation, and (or) reduction of vitamins, steroids, xenobiotics, and the majority of cardiovascular drugs in an oxygen- and NADPH-dependent manner. NADP 200-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 15513903-3 2004 Microsomal cytochrome P-450s induced production of reactive radical species, leading to differential modification of the proteins. reactive radical species 51-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 15513903-6 2004 Cytochrome P-450 inhibitors or antioxidants inhibited the production of reactive radical species and protein modification. reactive radical species 72-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 15585441-12 2004 As with other protease inhibitors, atazanavir is also a substrate and moderate inhibitor of the cytochrome P450 (CYP) system, in particular CYP3A4 and CYP2C9. Atazanavir Sulfate 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 15535984-1 2004 Two in vitro studies assessed the potential of daptomycin (Cubicin), a newly marketed antibiotic, to affect the cytochrome P450 (CYP450) isoforms in primary cultured human hepatocytes. Daptomycin 47-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-127 15535984-1 2004 Two in vitro studies assessed the potential of daptomycin (Cubicin), a newly marketed antibiotic, to affect the cytochrome P450 (CYP450) isoforms in primary cultured human hepatocytes. Daptomycin 47-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-135 15451555-0 2004 Non-specific inhibition of human cytochrome P450-catalyzed reactions by hemin. Hemin 72-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 15548707-2 2004 We found that ellipticine also forms the cytochrome P450 (CYP)-mediated covalent DNA adducts. ellipticine 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 15548707-2 2004 We found that ellipticine also forms the cytochrome P450 (CYP)-mediated covalent DNA adducts. ellipticine 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 15451555-2 2004 Inhibitory effects of hemin on the cytochrome P450 (CYP)-catalyzed reactions of human liver microsomes and reconstituted systems containing purified CYP and NADPH-cytochrome P450 reductase (NPR) were seen. Hemin 22-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 15451555-2 2004 Inhibitory effects of hemin on the cytochrome P450 (CYP)-catalyzed reactions of human liver microsomes and reconstituted systems containing purified CYP and NADPH-cytochrome P450 reductase (NPR) were seen. Hemin 22-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 15560776-2 2004 Cytochrome P450 enzymes promote a number of oxidative biotransformations including the hydroxylation of unactivated hydrocarbons. Hydrocarbons 116-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15451555-3 2004 Hemin non-specifically inhibited all of the microsomal CYP activities examined. Hemin 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 15451555-6 2004 Spectrally detectable CYP was destroyed in human liver microsomes and in a reconstituted system in the presence of hemin and an NADPH-generating system. Hemin 115-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 15451555-6 2004 Spectrally detectable CYP was destroyed in human liver microsomes and in a reconstituted system in the presence of hemin and an NADPH-generating system. NADP 128-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 15451555-7 2004 We propose that the antimutagenic effect of hemin might be due to inhibition of CYP and NPR enzymes involved in the bioactivation of mutagens. Hemin 44-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 15543094-7 2004 These proinflammatory cytokines down-regulate gene expression of major cytochrome P-450 and/or other enzymes with the specific effects on mRNA levels, protein expression, and enzyme activity, thus affecting metabolism of several endogenous lipophilic substances, such as steroids, lipid-soluble vitamins, prostaglandins, leukotrienes, thromboxanes, and exogenous substances. Steroids 271-279 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 15543094-7 2004 These proinflammatory cytokines down-regulate gene expression of major cytochrome P-450 and/or other enzymes with the specific effects on mRNA levels, protein expression, and enzyme activity, thus affecting metabolism of several endogenous lipophilic substances, such as steroids, lipid-soluble vitamins, prostaglandins, leukotrienes, thromboxanes, and exogenous substances. Prostaglandins 305-319 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 15543094-7 2004 These proinflammatory cytokines down-regulate gene expression of major cytochrome P-450 and/or other enzymes with the specific effects on mRNA levels, protein expression, and enzyme activity, thus affecting metabolism of several endogenous lipophilic substances, such as steroids, lipid-soluble vitamins, prostaglandins, leukotrienes, thromboxanes, and exogenous substances. Leukotrienes 321-333 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 15543094-7 2004 These proinflammatory cytokines down-regulate gene expression of major cytochrome P-450 and/or other enzymes with the specific effects on mRNA levels, protein expression, and enzyme activity, thus affecting metabolism of several endogenous lipophilic substances, such as steroids, lipid-soluble vitamins, prostaglandins, leukotrienes, thromboxanes, and exogenous substances. Thromboxanes 335-347 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 15372129-0 2004 Impact of cytochrome P-450 inhibition by cimetidine and induction by carbamazepine on the kinetics of hypericin and pseudohypericin in healthy volunteers. Cimetidine 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 15372129-0 2004 Impact of cytochrome P-450 inhibition by cimetidine and induction by carbamazepine on the kinetics of hypericin and pseudohypericin in healthy volunteers. hypericin 102-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. Nitrogen 88-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 15528841-2 2004 However, the information regarding human CYP inhibition by antituberculosis drugs is limited to isoniazid. Isoniazid 96-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 15528841-3 2004 In the current study, we examined the inhibitory effects of pyrazinamide and ethionamide, both of which are chemically related to isoniazid, on the CYP-mediated activities in human liver microsomes and compared them to that of isoniazid. Pyrazinamide 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 15528841-3 2004 In the current study, we examined the inhibitory effects of pyrazinamide and ethionamide, both of which are chemically related to isoniazid, on the CYP-mediated activities in human liver microsomes and compared them to that of isoniazid. Ethionamide 77-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 15528841-3 2004 In the current study, we examined the inhibitory effects of pyrazinamide and ethionamide, both of which are chemically related to isoniazid, on the CYP-mediated activities in human liver microsomes and compared them to that of isoniazid. Isoniazid 130-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 15528841-9 2004 In contrast, isoniazid might contribute to the severe drug interactions by a different inhibitory mechanism depending on each of the CYP isozymes, in addition to the reported observations. Isoniazid 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 15662082-1 2004 Although there are no literature data, which would clearly describe the role of catecholaminergic systems (including dopaminergic system) in the expression of the liver cytochrome P-450, there are well documented reports on physiological regulation of cytochrome P-450 by endogenous hormones (glucocorticoids, sex hormones, thyroid hormones, growth hormone, insulin) and by the immune system (cytokines), and all these factors remain under central nervous system control. Growth Hormone 342-356 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 15662082-1 2004 Although there are no literature data, which would clearly describe the role of catecholaminergic systems (including dopaminergic system) in the expression of the liver cytochrome P-450, there are well documented reports on physiological regulation of cytochrome P-450 by endogenous hormones (glucocorticoids, sex hormones, thyroid hormones, growth hormone, insulin) and by the immune system (cytokines), and all these factors remain under central nervous system control. Growth Hormone 342-356 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 252-268 15662082-3 2004 The mechanism underlying dopamine-induced regulation of the liver cytochrome P-450 expression seems not to be direct, but rather mediated by pituitary hormones and cytokines. Dopamine 25-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 15696928-1 2004 AIM: To identify the cytochrome P450 (CYP) isoform (s) involved in daidzein mono-hydroxylated metabolites using human liver microsomes. daidzein 67-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 15696928-1 2004 AIM: To identify the cytochrome P450 (CYP) isoform (s) involved in daidzein mono-hydroxylated metabolites using human liver microsomes. daidzein 67-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 15696928-3 2004 Nine selective inhibitors or substrate probes specific for different CYP isoforms were applied for screening the isoform(s) responsible for mono-hydroxylated metabolism of daidzein. daidzein 172-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. Nitrogen 88-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. Nitrogen 88-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 273-276 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. udenafil 130-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. udenafil 130-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. udenafil 130-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 273-276 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. DA-8164 141-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. DA-8164 141-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. DA-8164 141-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 273-276 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. udenafil 262-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 15801542-1 2004 The purpose of this paper was to characterize cytochrome P450 (CYP) enzymes involved in N-dealkylation of a new oral erectogenic, DA-8159 to DA-8164, a major circulating active metabolite, in human liver microsomes and to investigate the inhibitory potential of DA-8159 on CYP enzymes. udenafil 262-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 15801542-2 2004 CYP3A4 was identified as the major enzyme responsible for DA-8159 N-dealkylation to DA-8164 based on correlation analysis and specific CYP inhibitor and antibody-mediated inhibition study in human liver microsomes, and DA-8159 metabolism in cDNA expressed CYP enzymes. udenafil 58-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 15801542-2 2004 CYP3A4 was identified as the major enzyme responsible for DA-8159 N-dealkylation to DA-8164 based on correlation analysis and specific CYP inhibitor and antibody-mediated inhibition study in human liver microsomes, and DA-8159 metabolism in cDNA expressed CYP enzymes. udenafil 58-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 15801542-2 2004 CYP3A4 was identified as the major enzyme responsible for DA-8159 N-dealkylation to DA-8164 based on correlation analysis and specific CYP inhibitor and antibody-mediated inhibition study in human liver microsomes, and DA-8159 metabolism in cDNA expressed CYP enzymes. DA-8164 84-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 15801542-2 2004 CYP3A4 was identified as the major enzyme responsible for DA-8159 N-dealkylation to DA-8164 based on correlation analysis and specific CYP inhibitor and antibody-mediated inhibition study in human liver microsomes, and DA-8159 metabolism in cDNA expressed CYP enzymes. udenafil 219-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 15801544-1 2004 The study was carried out to identify and characterize kinetically the cytochrome P450 (CYP) enzymes responsible for the major metabolite formation of quazepam. quazepam 151-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 15801544-1 2004 The study was carried out to identify and characterize kinetically the cytochrome P450 (CYP) enzymes responsible for the major metabolite formation of quazepam. quazepam 151-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 15382119-2 2004 At the hepatic level, the orphan nuclear constitutive androstane receptor (CAR) (NR1I3) controls phase I (cytochrome P450 [CYP] 2B and CYP3A), phase II (UGT1A1), and transporter (SLC21A6, MRP2) genes involved in drug metabolism and bilirubin clearance in response to xenobiotics such as phenobarbital or endobiotics such as bilirubin. Bilirubin 324-333 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-130 15554232-2 2004 Upon application of this method to patients receiving different kinds of drug therapy, Herbert Remmer was the first to describe that total human hepatic cytochrome P450 was markedly elevated by the new anti-tuberculosis drug rifampicin. Rifampin 225-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-168 15382119-2 2004 At the hepatic level, the orphan nuclear constitutive androstane receptor (CAR) (NR1I3) controls phase I (cytochrome P450 [CYP] 2B and CYP3A), phase II (UGT1A1), and transporter (SLC21A6, MRP2) genes involved in drug metabolism and bilirubin clearance in response to xenobiotics such as phenobarbital or endobiotics such as bilirubin. Bilirubin 232-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-130 15356430-8 2004 Because several inflammatory cytokines down-regulate gene expression of major cytochrome P-450 and/or other enzymes with the specific effects on mRNA levels, protein expression, and enzyme activity, thus affecting the metabolism of several endogenous lipophilic substances such as steroids, lipid-soluble vitamins, prostaglandins, leukotrienes, thromboxanes, and exogenous substances, their irregularities in the body may eventually lead to the flare of latent diseases in some predisposed subjects. Steroids 281-289 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 15382119-2 2004 At the hepatic level, the orphan nuclear constitutive androstane receptor (CAR) (NR1I3) controls phase I (cytochrome P450 [CYP] 2B and CYP3A), phase II (UGT1A1), and transporter (SLC21A6, MRP2) genes involved in drug metabolism and bilirubin clearance in response to xenobiotics such as phenobarbital or endobiotics such as bilirubin. Phenobarbital 287-300 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-130 15764407-0 2004 Biotransformation of chlorpyrifos and diazinon by human liver microsomes and recombinant human cytochrome P450s (CYP). Chlorpyrifos 21-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 15764407-0 2004 Biotransformation of chlorpyrifos and diazinon by human liver microsomes and recombinant human cytochrome P450s (CYP). Diazinon 38-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 15764407-1 2004 The cytochrome P450 (CYP)-mediated biotransformation of the organophosphorothioate insecticides chlorpyrifos and diazinon was investigated. organophosphorothioate 60-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 15764407-1 2004 The cytochrome P450 (CYP)-mediated biotransformation of the organophosphorothioate insecticides chlorpyrifos and diazinon was investigated. organophosphorothioate 60-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 15764407-1 2004 The cytochrome P450 (CYP)-mediated biotransformation of the organophosphorothioate insecticides chlorpyrifos and diazinon was investigated. Chlorpyrifos 96-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 15764407-1 2004 The cytochrome P450 (CYP)-mediated biotransformation of the organophosphorothioate insecticides chlorpyrifos and diazinon was investigated. Chlorpyrifos 96-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 15764407-1 2004 The cytochrome P450 (CYP)-mediated biotransformation of the organophosphorothioate insecticides chlorpyrifos and diazinon was investigated. Diazinon 113-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 15764407-1 2004 The cytochrome P450 (CYP)-mediated biotransformation of the organophosphorothioate insecticides chlorpyrifos and diazinon was investigated. Diazinon 113-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 15764407-9 2004 However, the role of individual CYP enzymes in these two biotransformation pathways varied according to the structure of the organophosphorothioate, which was reflected in different activation/detoxification ratios for chlorpyrifos and diazinon. organophosphorothioate 125-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 15764407-9 2004 However, the role of individual CYP enzymes in these two biotransformation pathways varied according to the structure of the organophosphorothioate, which was reflected in different activation/detoxification ratios for chlorpyrifos and diazinon. Chlorpyrifos 219-231 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 15764407-9 2004 However, the role of individual CYP enzymes in these two biotransformation pathways varied according to the structure of the organophosphorothioate, which was reflected in different activation/detoxification ratios for chlorpyrifos and diazinon. Diazinon 236-244 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 15764407-10 2004 Variability in activity of individual CYP enzymes may influence interindividual sensitivity to the toxic effects of chlorpyrifos and diazinon. Chlorpyrifos 116-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 15258138-0 2004 Radical rebound mechanism in cytochrome P-450-catalyzed hydroxylation of the multifaceted radical clocks alpha- and beta-thujone. alpha- and beta-thujone 105-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 15764407-10 2004 Variability in activity of individual CYP enzymes may influence interindividual sensitivity to the toxic effects of chlorpyrifos and diazinon. Diazinon 133-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 15356430-8 2004 Because several inflammatory cytokines down-regulate gene expression of major cytochrome P-450 and/or other enzymes with the specific effects on mRNA levels, protein expression, and enzyme activity, thus affecting the metabolism of several endogenous lipophilic substances such as steroids, lipid-soluble vitamins, prostaglandins, leukotrienes, thromboxanes, and exogenous substances, their irregularities in the body may eventually lead to the flare of latent diseases in some predisposed subjects. Prostaglandins 315-329 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 15356430-8 2004 Because several inflammatory cytokines down-regulate gene expression of major cytochrome P-450 and/or other enzymes with the specific effects on mRNA levels, protein expression, and enzyme activity, thus affecting the metabolism of several endogenous lipophilic substances such as steroids, lipid-soluble vitamins, prostaglandins, leukotrienes, thromboxanes, and exogenous substances, their irregularities in the body may eventually lead to the flare of latent diseases in some predisposed subjects. Leukotrienes 331-343 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 15356430-8 2004 Because several inflammatory cytokines down-regulate gene expression of major cytochrome P-450 and/or other enzymes with the specific effects on mRNA levels, protein expression, and enzyme activity, thus affecting the metabolism of several endogenous lipophilic substances such as steroids, lipid-soluble vitamins, prostaglandins, leukotrienes, thromboxanes, and exogenous substances, their irregularities in the body may eventually lead to the flare of latent diseases in some predisposed subjects. Thromboxanes 345-357 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 15503650-4 2004 Cytochrome P450 metabolism of arachidonic acid to 20-hydroxyeicosatetraenoic acid (20-HETE) or epoxyeicosatrienoic acids (EETs) provides a mechanism for the constriction and relaxation of cerebral arteries, respectively. Arachidonic Acid 30-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15314576-1 2004 The porphyrin complexes of manganese catalysts are biomimetic catalysts whose structural analogy with the active site of cytochrome P-450 enzymes can be used to obtain synthetic models of therapeutic agent metabolites. Porphyrins 4-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 15314576-1 2004 The porphyrin complexes of manganese catalysts are biomimetic catalysts whose structural analogy with the active site of cytochrome P-450 enzymes can be used to obtain synthetic models of therapeutic agent metabolites. Manganese 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 15503650-4 2004 Cytochrome P450 metabolism of arachidonic acid to 20-hydroxyeicosatetraenoic acid (20-HETE) or epoxyeicosatrienoic acids (EETs) provides a mechanism for the constriction and relaxation of cerebral arteries, respectively. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 50-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15503650-4 2004 Cytochrome P450 metabolism of arachidonic acid to 20-hydroxyeicosatetraenoic acid (20-HETE) or epoxyeicosatrienoic acids (EETs) provides a mechanism for the constriction and relaxation of cerebral arteries, respectively. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 83-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15503650-4 2004 Cytochrome P450 metabolism of arachidonic acid to 20-hydroxyeicosatetraenoic acid (20-HETE) or epoxyeicosatrienoic acids (EETs) provides a mechanism for the constriction and relaxation of cerebral arteries, respectively. epoxyeicosatrienoic acids 95-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15503650-4 2004 Cytochrome P450 metabolism of arachidonic acid to 20-hydroxyeicosatetraenoic acid (20-HETE) or epoxyeicosatrienoic acids (EETs) provides a mechanism for the constriction and relaxation of cerebral arteries, respectively. eets 122-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15669763-0 2004 Biomimetic oxidation of unactivated carbons in steroids by a model of cytochrome P-450, oxorutheniumporphyrinate complex. Carbon 36-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 15669763-0 2004 Biomimetic oxidation of unactivated carbons in steroids by a model of cytochrome P-450, oxorutheniumporphyrinate complex. Steroids 47-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 15669763-0 2004 Biomimetic oxidation of unactivated carbons in steroids by a model of cytochrome P-450, oxorutheniumporphyrinate complex. oxorutheniumporphyrinate 88-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 15669763-1 2004 Biomimetic oxidation of unactivated carbons for structurally different steroids was studied with a model of cytochrome P-450, oxorutheniumporphyrinate complex, which is generated in situ by 2,6-dichloropyridine N-oxide as an oxygen donor and (5,10,15,20-tetramesitylporphyrinate) ruthenium(II) carbonyl complex and HBr as catalysts. Carbon 36-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 15669763-1 2004 Biomimetic oxidation of unactivated carbons for structurally different steroids was studied with a model of cytochrome P-450, oxorutheniumporphyrinate complex, which is generated in situ by 2,6-dichloropyridine N-oxide as an oxygen donor and (5,10,15,20-tetramesitylporphyrinate) ruthenium(II) carbonyl complex and HBr as catalysts. Steroids 71-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 15669763-1 2004 Biomimetic oxidation of unactivated carbons for structurally different steroids was studied with a model of cytochrome P-450, oxorutheniumporphyrinate complex, which is generated in situ by 2,6-dichloropyridine N-oxide as an oxygen donor and (5,10,15,20-tetramesitylporphyrinate) ruthenium(II) carbonyl complex and HBr as catalysts. oxorutheniumporphyrinate 126-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 15669763-1 2004 Biomimetic oxidation of unactivated carbons for structurally different steroids was studied with a model of cytochrome P-450, oxorutheniumporphyrinate complex, which is generated in situ by 2,6-dichloropyridine N-oxide as an oxygen donor and (5,10,15,20-tetramesitylporphyrinate) ruthenium(II) carbonyl complex and HBr as catalysts. 2,6-dichloropyridine N-oxide 190-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 15669763-1 2004 Biomimetic oxidation of unactivated carbons for structurally different steroids was studied with a model of cytochrome P-450, oxorutheniumporphyrinate complex, which is generated in situ by 2,6-dichloropyridine N-oxide as an oxygen donor and (5,10,15,20-tetramesitylporphyrinate) ruthenium(II) carbonyl complex and HBr as catalysts. Oxygen 225-231 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 15669763-1 2004 Biomimetic oxidation of unactivated carbons for structurally different steroids was studied with a model of cytochrome P-450, oxorutheniumporphyrinate complex, which is generated in situ by 2,6-dichloropyridine N-oxide as an oxygen donor and (5,10,15,20-tetramesitylporphyrinate) ruthenium(II) carbonyl complex and HBr as catalysts. (5,10,15,20-tetramesitylporphyrinate) ruthenium(ii) carbonyl complex 242-310 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 15669763-1 2004 Biomimetic oxidation of unactivated carbons for structurally different steroids was studied with a model of cytochrome P-450, oxorutheniumporphyrinate complex, which is generated in situ by 2,6-dichloropyridine N-oxide as an oxygen donor and (5,10,15,20-tetramesitylporphyrinate) ruthenium(II) carbonyl complex and HBr as catalysts. Hydrobromic Acid 315-318 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 15247556-0 2004 Coadministration of lopinavir/ritonavir and phenytoin results in two-way drug interaction through cytochrome P-450 induction. Lopinavir 20-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 15315710-0 2004 Inducible cytochrome P450 activities in renal glomerular mesangial cells: biochemical basis for antagonistic interactions among nephrocarcinogenic polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 147-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 15159443-0 2004 Comprehensive evaluation of tamoxifen sequential biotransformation by the human cytochrome P450 system in vitro: prominent roles for CYP3A and CYP2D6. Tamoxifen 28-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 15276085-1 2004 This study was aimed at identifying the isoform(s) of human liver cytochrome P450 (CYP) involved in the hepatic biotransformation of trans-resveratrol (trans-3,5,4"-trihydroxystilbene). Resveratrol 133-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 15276085-1 2004 This study was aimed at identifying the isoform(s) of human liver cytochrome P450 (CYP) involved in the hepatic biotransformation of trans-resveratrol (trans-3,5,4"-trihydroxystilbene). Resveratrol 133-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 15276085-1 2004 This study was aimed at identifying the isoform(s) of human liver cytochrome P450 (CYP) involved in the hepatic biotransformation of trans-resveratrol (trans-3,5,4"-trihydroxystilbene). trans-3,5,4"-trihydroxystilbene 152-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 15276085-1 2004 This study was aimed at identifying the isoform(s) of human liver cytochrome P450 (CYP) involved in the hepatic biotransformation of trans-resveratrol (trans-3,5,4"-trihydroxystilbene). trans-3,5,4"-trihydroxystilbene 152-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 15247556-0 2004 Coadministration of lopinavir/ritonavir and phenytoin results in two-way drug interaction through cytochrome P-450 induction. Ritonavir 30-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 15247556-0 2004 Coadministration of lopinavir/ritonavir and phenytoin results in two-way drug interaction through cytochrome P-450 induction. Phenytoin 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 15247556-1 2004 Lopinavir/ritonavir (LPV/RTV) is a CYP3A4 inhibitor and substrate; it also may induce cytochrome P-450 (CYP) isozymes. Lopinavir 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 15247556-1 2004 Lopinavir/ritonavir (LPV/RTV) is a CYP3A4 inhibitor and substrate; it also may induce cytochrome P-450 (CYP) isozymes. Lopinavir 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 15247556-1 2004 Lopinavir/ritonavir (LPV/RTV) is a CYP3A4 inhibitor and substrate; it also may induce cytochrome P-450 (CYP) isozymes. Ritonavir 10-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 15247556-1 2004 Lopinavir/ritonavir (LPV/RTV) is a CYP3A4 inhibitor and substrate; it also may induce cytochrome P-450 (CYP) isozymes. Ritonavir 10-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 15122071-2 2004 To assess the biotransformation and the potential implications for human pharmacology and toxicology, the in vitro metabolism of thiocoraline was characterized using human plasma, human liver preparations, cytochrome P450 (CYP) and uridine diphosphoglucuronosyl transferase (UGT) supersomes and human cell lines. thiocoraline 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-221 15289791-0 2004 Human sympathetic activation by alpha2-adrenergic blockade with yohimbine: Bimodal, epistatic influence of cytochrome P450-mediated drug metabolism. Yohimbine 64-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 15122071-2 2004 To assess the biotransformation and the potential implications for human pharmacology and toxicology, the in vitro metabolism of thiocoraline was characterized using human plasma, human liver preparations, cytochrome P450 (CYP) and uridine diphosphoglucuronosyl transferase (UGT) supersomes and human cell lines. thiocoraline 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-226 15122071-4 2004 Using CYP supersomes it was shown that thiocoraline is mainly metabolized by CYP3A4, with CYP1A1, CYP2C8 and CYP2C9 playing a minor role in the biotransformation (<3%). thiocoraline 39-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 15122071-10 2004 These results provide evidence that human CYP3A4 plays a major role in the metabolism of thiocoraline in vitro and that the metabolites formed by CYP are conjugated by the phase II enzymes UGT, ST and GST. thiocoraline 89-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 15145985-0 2004 Human CYP4F3s are the main catalysts in the oxidation of fatty acid epoxides. fatty acid epoxides 57-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-12 15292328-1 2004 Sequential conversion of estradiol to catecholestradiols and methoxyestradiols by cytochrome-P(450) (CYP450) and catechol-O-methyltransferase (COMT), respectively, contributes to the antimitogenic effects of estradiol on vascular smooth muscle cell (SMC) growth via estrogen receptor-independent mechanisms. Estradiol 25-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-99 15292328-1 2004 Sequential conversion of estradiol to catecholestradiols and methoxyestradiols by cytochrome-P(450) (CYP450) and catechol-O-methyltransferase (COMT), respectively, contributes to the antimitogenic effects of estradiol on vascular smooth muscle cell (SMC) growth via estrogen receptor-independent mechanisms. Estradiol 25-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 15292328-1 2004 Sequential conversion of estradiol to catecholestradiols and methoxyestradiols by cytochrome-P(450) (CYP450) and catechol-O-methyltransferase (COMT), respectively, contributes to the antimitogenic effects of estradiol on vascular smooth muscle cell (SMC) growth via estrogen receptor-independent mechanisms. catecholestradiols 38-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-99 15292328-1 2004 Sequential conversion of estradiol to catecholestradiols and methoxyestradiols by cytochrome-P(450) (CYP450) and catechol-O-methyltransferase (COMT), respectively, contributes to the antimitogenic effects of estradiol on vascular smooth muscle cell (SMC) growth via estrogen receptor-independent mechanisms. catecholestradiols 38-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 15292328-1 2004 Sequential conversion of estradiol to catecholestradiols and methoxyestradiols by cytochrome-P(450) (CYP450) and catechol-O-methyltransferase (COMT), respectively, contributes to the antimitogenic effects of estradiol on vascular smooth muscle cell (SMC) growth via estrogen receptor-independent mechanisms. methoxyestradiols 61-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-99 15292328-1 2004 Sequential conversion of estradiol to catecholestradiols and methoxyestradiols by cytochrome-P(450) (CYP450) and catechol-O-methyltransferase (COMT), respectively, contributes to the antimitogenic effects of estradiol on vascular smooth muscle cell (SMC) growth via estrogen receptor-independent mechanisms. methoxyestradiols 61-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 15292328-1 2004 Sequential conversion of estradiol to catecholestradiols and methoxyestradiols by cytochrome-P(450) (CYP450) and catechol-O-methyltransferase (COMT), respectively, contributes to the antimitogenic effects of estradiol on vascular smooth muscle cell (SMC) growth via estrogen receptor-independent mechanisms. Estradiol 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-99 15292328-1 2004 Sequential conversion of estradiol to catecholestradiols and methoxyestradiols by cytochrome-P(450) (CYP450) and catechol-O-methyltransferase (COMT), respectively, contributes to the antimitogenic effects of estradiol on vascular smooth muscle cell (SMC) growth via estrogen receptor-independent mechanisms. Estradiol 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-107 15145985-8 2004 Moreover, vicinal diol from both C18-epoxides and EETs were omega-hydroxylated by liver microsomes and by CYP4F2 and CYP4F3. vicinal diol 10-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-123 15145985-8 2004 Moreover, vicinal diol from both C18-epoxides and EETs were omega-hydroxylated by liver microsomes and by CYP4F2 and CYP4F3. c18-epoxides 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-123 15145985-8 2004 Moreover, vicinal diol from both C18-epoxides and EETs were omega-hydroxylated by liver microsomes and by CYP4F2 and CYP4F3. eets 50-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-123 15285849-7 2004 However, flavonol aglycones, the biflavonol amentoflavone and several other non-glycosidic constituents are significant in-vitro inhibitors of CYP. 3-hydroxyflavone 9-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-146 15285849-7 2004 However, flavonol aglycones, the biflavonol amentoflavone and several other non-glycosidic constituents are significant in-vitro inhibitors of CYP. aglycones 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-146 15285849-7 2004 However, flavonol aglycones, the biflavonol amentoflavone and several other non-glycosidic constituents are significant in-vitro inhibitors of CYP. biflavonol amentoflavone 33-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-146 15672756-2 2004 It has previously been reported that N,N-dipropyl-2-[4-methoxy-3-(2-phenylethoxy)phenyl]-ethylamine monohydrochloride (NE-100) was predominantly metabolized by cytochrome P450 (CYP) 2D6 in human liver microsomes (HLM). N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 37-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-175 15262846-1 2004 BACKGROUND: 20-hydroxyeicosatetraenoic acid (20-HETE) is a cytochrome P450 (omega-hydroxylase) metabolite of arachidonic acid with vasoconstrictor activity that may be involved in the pathogenesis of hypertension. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 12-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-93 15262846-1 2004 BACKGROUND: 20-hydroxyeicosatetraenoic acid (20-HETE) is a cytochrome P450 (omega-hydroxylase) metabolite of arachidonic acid with vasoconstrictor activity that may be involved in the pathogenesis of hypertension. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 45-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-93 15262846-1 2004 BACKGROUND: 20-hydroxyeicosatetraenoic acid (20-HETE) is a cytochrome P450 (omega-hydroxylase) metabolite of arachidonic acid with vasoconstrictor activity that may be involved in the pathogenesis of hypertension. Arachidonic Acid 109-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-93 15238214-4 2004 The HL60 cells, which were differentiated into PMN-like shapes by treatment with all-trans-retinoic acid (RA), also expressed CYP4F3, CYP4F3B and CYP4F12. Tretinoin 81-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-132 15238214-4 2004 The HL60 cells, which were differentiated into PMN-like shapes by treatment with all-trans-retinoic acid (RA), also expressed CYP4F3, CYP4F3B and CYP4F12. Tretinoin 106-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-132 15238214-5 2004 CYP4F3 was expressed in one third of the peripheral monocytes, which omega-hydroxylated leukotriene B(4) (LTB(4)) at a rate 11 times lower than that of PMN. Leukotriene B4 88-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 15238214-5 2004 CYP4F3 was expressed in one third of the peripheral monocytes, which omega-hydroxylated leukotriene B(4) (LTB(4)) at a rate 11 times lower than that of PMN. Leukotriene B4 106-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 15238214-6 2004 The cells that were differentiated into a form similar to monocytes/macrophages in shape by treatment with 12-myristate 13-acetate expressed mRNA for CYP4F3 and CYP4F3B. 12-myristate 13-acetate 107-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-156 15248218-0 2004 Cytochrome P450 pharmacogenetics as a predictor of toxicity and clinical response to pulse cyclophosphamide in lupus nephritis. Cyclophosphamide 91-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15248218-3 2004 Cyclophosphamide is a prodrug that requires activation by cytochrome P450 (CYP) enzymes. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 15248218-3 2004 Cyclophosphamide is a prodrug that requires activation by cytochrome P450 (CYP) enzymes. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 15248218-10 2004 CONCLUSION: Determination of selected cytochrome P450 enzyme genotypes may be valuable for predicting the risk of premature ovarian failure in lupus nephritis patients treated with cyclophosphamide. Cyclophosphamide 181-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 15197521-1 2004 OBJECTIVE: This study aimed to investigate the importance of cytochrome P(450) enzymes for the reported interaction between tramadol and warfarin. Tramadol 124-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-78 15197521-1 2004 OBJECTIVE: This study aimed to investigate the importance of cytochrome P(450) enzymes for the reported interaction between tramadol and warfarin. Warfarin 137-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-78 15537169-1 2004 The human cytochrome P450 (CYP) isoforms catalyzing the oxidation metabolism of desglymidodrine (DMAE), an active metabolite of midodrine, were studied. desglymidodrine 80-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 15537169-1 2004 The human cytochrome P450 (CYP) isoforms catalyzing the oxidation metabolism of desglymidodrine (DMAE), an active metabolite of midodrine, were studied. desglymidodrine 80-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 15537169-1 2004 The human cytochrome P450 (CYP) isoforms catalyzing the oxidation metabolism of desglymidodrine (DMAE), an active metabolite of midodrine, were studied. Deanol 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 15537169-1 2004 The human cytochrome P450 (CYP) isoforms catalyzing the oxidation metabolism of desglymidodrine (DMAE), an active metabolite of midodrine, were studied. Deanol 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 15537169-1 2004 The human cytochrome P450 (CYP) isoforms catalyzing the oxidation metabolism of desglymidodrine (DMAE), an active metabolite of midodrine, were studied. Midodrine 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 15537169-1 2004 The human cytochrome P450 (CYP) isoforms catalyzing the oxidation metabolism of desglymidodrine (DMAE), an active metabolite of midodrine, were studied. Midodrine 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 15537169-4 2004 Quinidine (a selective inhibitor of CYP2D6) inhibited the O-demethylation of DMAE in pooled human microsomes by 86%, while selective inhibitors for other forms of CYP did not show any appreciable effect. Quinidine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 15537169-4 2004 Quinidine (a selective inhibitor of CYP2D6) inhibited the O-demethylation of DMAE in pooled human microsomes by 86%, while selective inhibitors for other forms of CYP did not show any appreciable effect. Deanol 77-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 15672756-2 2004 It has previously been reported that N,N-dipropyl-2-[4-methoxy-3-(2-phenylethoxy)phenyl]-ethylamine monohydrochloride (NE-100) was predominantly metabolized by cytochrome P450 (CYP) 2D6 in human liver microsomes (HLM). N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 37-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 15672756-3 2004 In the present study, the contribution of CYP forms involved in the formation of the major metabolites of NE-100 in human liver lacking CYP2D6 activity (PM-HLM) has been predicted by use of in vitro kinetic data on recombinant CYPs microsomes (rCYPs). N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 106-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 15081936-3 2004 Cytochrome P450 dependent epoxidation and non-enzymic lipid peroxidation of oleic acid (cis-9-octadecenoic acid) result in the formation of cis-9,10-epoxyoctadecanoic acid (cis-EODA). 9,10-epoxystearic acid 140-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 14751845-2 2004 We also explored the contribution of cytochrome P-450 (CYP450) product of arachidonic acid (AA) metabolism, hydrogen peroxide (H(2)O(2)), and gap junctions that have been suggested to be involved in EDHF-mediated responses. Arachidonic Acid 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 14751845-2 2004 We also explored the contribution of cytochrome P-450 (CYP450) product of arachidonic acid (AA) metabolism, hydrogen peroxide (H(2)O(2)), and gap junctions that have been suggested to be involved in EDHF-mediated responses. Arachidonic Acid 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-61 20945304-0 2004 Human cytochrome P450: metabolism of testosterone by CYP3A4 and inhibition by ketoconazole. Testosterone 37-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 20945304-0 2004 Human cytochrome P450: metabolism of testosterone by CYP3A4 and inhibition by ketoconazole. Ketoconazole 78-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 15204103-0 2004 In vitro evaluation of cytochrome P450-mediated drug interactions between cytarabine, idarubicin, itraconazole and caspofungin. Cytarabine 74-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 15204103-0 2004 In vitro evaluation of cytochrome P450-mediated drug interactions between cytarabine, idarubicin, itraconazole and caspofungin. Idarubicin 86-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 15204103-0 2004 In vitro evaluation of cytochrome P450-mediated drug interactions between cytarabine, idarubicin, itraconazole and caspofungin. Itraconazole 98-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 15204103-0 2004 In vitro evaluation of cytochrome P450-mediated drug interactions between cytarabine, idarubicin, itraconazole and caspofungin. Caspofungin 115-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 15204103-3 2004 In vitro assays were performed to assess whether cytochrome P450 (CYP450) enzymes were affected by combinations of cytarabine or idarubicin with itraconazole or caspofungin. Cytarabine 115-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 15204103-3 2004 In vitro assays were performed to assess whether cytochrome P450 (CYP450) enzymes were affected by combinations of cytarabine or idarubicin with itraconazole or caspofungin. Cytarabine 115-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-72 15204103-3 2004 In vitro assays were performed to assess whether cytochrome P450 (CYP450) enzymes were affected by combinations of cytarabine or idarubicin with itraconazole or caspofungin. Idarubicin 129-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 15204103-3 2004 In vitro assays were performed to assess whether cytochrome P450 (CYP450) enzymes were affected by combinations of cytarabine or idarubicin with itraconazole or caspofungin. Idarubicin 129-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-72 15204103-3 2004 In vitro assays were performed to assess whether cytochrome P450 (CYP450) enzymes were affected by combinations of cytarabine or idarubicin with itraconazole or caspofungin. Itraconazole 145-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 15204103-3 2004 In vitro assays were performed to assess whether cytochrome P450 (CYP450) enzymes were affected by combinations of cytarabine or idarubicin with itraconazole or caspofungin. Itraconazole 145-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-72 15204103-3 2004 In vitro assays were performed to assess whether cytochrome P450 (CYP450) enzymes were affected by combinations of cytarabine or idarubicin with itraconazole or caspofungin. Caspofungin 161-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 15204103-3 2004 In vitro assays were performed to assess whether cytochrome P450 (CYP450) enzymes were affected by combinations of cytarabine or idarubicin with itraconazole or caspofungin. Caspofungin 161-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-72 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Cytarabine 76-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-142 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Cytarabine 76-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-185 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Cytarabine 76-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-185 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Idarubicin 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-142 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Idarubicin 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-185 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Idarubicin 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-185 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Itraconazole 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-142 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Itraconazole 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-185 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Itraconazole 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-185 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Caspofungin 119-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-142 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Caspofungin 119-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-185 15204103-4 2004 METHODS: The high throughput microtiter assay was used to determine whether cytarabine, idarubicin and itraconazole or caspofungin were CYP450 isoenzyme substrates, inhibitors of CYP450 isoenzymes, and to determine potential CYP450 metabolism interactions between these agents. Caspofungin 119-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-185 15204103-5 2004 RESULTS: Idarubicin is a substrate for CYP450 2D6 and 2C9. Idarubicin 9-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-45 15204103-6 2004 Cytarabine is a substrate of CYP450 3A4. Cytarabine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-35 15204103-7 2004 Idarubicin inhibits CYP450 2D6, and cytarabine, itraconazole, and caspofungin inhibit CYP450 3A4. Idarubicin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-26 15204103-7 2004 Idarubicin inhibits CYP450 2D6, and cytarabine, itraconazole, and caspofungin inhibit CYP450 3A4. Cytarabine 36-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-92 15204103-7 2004 Idarubicin inhibits CYP450 2D6, and cytarabine, itraconazole, and caspofungin inhibit CYP450 3A4. Caspofungin 66-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-92 15046773-2 2004 OTA is metabolised by cytochrome P450 (CYP450) enzymes resulting in hydroxylated metabolites, 4R-OH-OTA and 4S-OH-OTA, and possibly in other minor metabolites including OTA-quinones. 4r-oh-ota 94-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 15046773-2 2004 OTA is metabolised by cytochrome P450 (CYP450) enzymes resulting in hydroxylated metabolites, 4R-OH-OTA and 4S-OH-OTA, and possibly in other minor metabolites including OTA-quinones. 4r-oh-ota 94-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-45 15046773-2 2004 OTA is metabolised by cytochrome P450 (CYP450) enzymes resulting in hydroxylated metabolites, 4R-OH-OTA and 4S-OH-OTA, and possibly in other minor metabolites including OTA-quinones. 4s-oh-ota 108-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 15046773-2 2004 OTA is metabolised by cytochrome P450 (CYP450) enzymes resulting in hydroxylated metabolites, 4R-OH-OTA and 4S-OH-OTA, and possibly in other minor metabolites including OTA-quinones. 4s-oh-ota 108-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-45 15046773-2 2004 OTA is metabolised by cytochrome P450 (CYP450) enzymes resulting in hydroxylated metabolites, 4R-OH-OTA and 4S-OH-OTA, and possibly in other minor metabolites including OTA-quinones. ota-quinones 169-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 15046773-2 2004 OTA is metabolised by cytochrome P450 (CYP450) enzymes resulting in hydroxylated metabolites, 4R-OH-OTA and 4S-OH-OTA, and possibly in other minor metabolites including OTA-quinones. ota-quinones 169-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-45 15180494-0 2004 Role of cytochrome P450 metabolites of arachidonic acid in hypertension. Arachidonic Acid 39-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 15180494-1 2004 Considerable evidence has accumulated over the last decade implicating a role of cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA) in the pathogenesis of hypertension. Arachidonic Acid 128-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 15180494-1 2004 Considerable evidence has accumulated over the last decade implicating a role of cytochrome P450 (CYP)-dependent metabolites of arachidonic acid (AA) in the pathogenesis of hypertension. Arachidonic Acid 128-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 15311563-4 2004 Posaconazole is a CYP3A4 inhibitor, but it does not inhibit the activity of other CYP enzymes. posaconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 15081936-3 2004 Cytochrome P450 dependent epoxidation and non-enzymic lipid peroxidation of oleic acid (cis-9-octadecenoic acid) result in the formation of cis-9,10-epoxyoctadecanoic acid (cis-EODA). 9,10-epoxystearic acid 173-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15081398-0 2004 Cytochrome P450-dependent toxicity of environmental polycyclic aromatic hydrocarbons towards human macrophages. Polycyclic Aromatic Hydrocarbons 52-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15128933-1 2004 The synthesis of bioactive vitamin D requires hydroxylation at the 1 alpha and 25 positions by cytochrome P450 enzymes in the kidney and liver, respectively. Vitamin D 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 15081421-5 2004 Especially, the heme-thiolate containing proteins cytochrome P450(BM-3) (wild type and F87Y variant) and prostacyclin synthase were nitrated with high efficacy. heme-thiolate 16-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-70 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. Tamoxifen 59-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-204 15156965-6 2004 Gefitinib is metabolized by the cytochrome P-450 isoenzyme system and may be affected by other drugs that influence this enzyme activity. Gefitinib 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 14764421-5 2004 Under steady-state conditions, most of the de novo synthesis of cholesterol in the brain appears to be balanced by excretion of the cytochrome P-450-generated oxysterol 24S-hydroxycholesterol. Oxysterols 159-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 14764421-5 2004 Under steady-state conditions, most of the de novo synthesis of cholesterol in the brain appears to be balanced by excretion of the cytochrome P-450-generated oxysterol 24S-hydroxycholesterol. 24-hydroxycholesterol 169-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. Tamoxifen 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-209 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. Tamoxifen 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-204 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. Tamoxifen 59-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-209 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. Tamoxifen 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-209 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. 4-hydroxy-N-desmethyltamoxifen 331-340 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-204 15237850-1 2004 The cytochrome P450 (P450) field came out of interest in the metabolism of drugs, carcinogens, and steroids, which remain major focal points. Steroids 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. 4-hydroxy-N-desmethyltamoxifen 80-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-204 15237852-7 2004 Furthermore, the results provide proof of principle for NMR as a method for studying CYP allosterism, and demonstrate that the model ligand 9-aminophenanthrene binds in two discrete events to individual subsites within the active site of CYP(eryF). 9-aminophenanthrene 140-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. 4-hydroxy-N-desmethyltamoxifen 331-340 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-209 15237852-7 2004 Furthermore, the results provide proof of principle for NMR as a method for studying CYP allosterism, and demonstrate that the model ligand 9-aminophenanthrene binds in two discrete events to individual subsites within the active site of CYP(eryF). 9-aminophenanthrene 140-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 238-241 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. 4-hydroxy-N-desmethyltamoxifen 80-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-209 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. 4-hydroxy-N-desmethyltamoxifen 115-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-204 15237857-1 2004 Cytochrome P450 (CYP) enzymes in the brain may have a role in the activation or inactivation of centrally acting drugs, in the metabolism of endogenous compounds, and in the generation of damaging toxic metabolites and/or oxygen stress. Oxygen 222-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15237857-1 2004 Cytochrome P450 (CYP) enzymes in the brain may have a role in the activation or inactivation of centrally acting drugs, in the metabolism of endogenous compounds, and in the generation of damaging toxic metabolites and/or oxygen stress. Oxygen 222-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. 4-hydroxy-N-desmethyltamoxifen 115-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 206-209 15111773-4 2004 A recent study has shown that plasma concentrations of the tamoxifen metabolite 4-hydroxy- N -desmethyl tamoxifen (endoxifen), in patents undergoing tamoxifen therapy, are dependent on the cytochrome p450 (CYP) 206 ge notype of the patient and that medications commonly prescribed to patients on tamoxifen therapy can also inhibit endoxifen production. Tamoxifen 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-204 15078344-1 2004 It has been shown that large doses of acetaminophen can result in increased degradation of the hepatic cytochrome P450 (CYP) enzymes in vivo; however, the proteolytic pathways have not been identified. Acetaminophen 38-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 15043991-0 2004 Inhibition of cytochrome P450 activities by oleanolic acid and ursolic acid in human liver microsomes. Oleanolic Acid 44-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 15043991-0 2004 Inhibition of cytochrome P450 activities by oleanolic acid and ursolic acid in human liver microsomes. ursolic acid 63-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 15056479-0 2004 The metabolism of the piperazine-type phenothiazine neuroleptic perazine by the human cytochrome P-450 isoenzymes. Piperazine 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 15056479-0 2004 The metabolism of the piperazine-type phenothiazine neuroleptic perazine by the human cytochrome P-450 isoenzymes. phenothiazine 38-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 15056479-0 2004 The metabolism of the piperazine-type phenothiazine neuroleptic perazine by the human cytochrome P-450 isoenzymes. Perazine 24-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 15056479-1 2004 Identification of cytochrome P-450 isoenzymes (CYPs) involved in perazine 5-sulphoxidation and N-demethylation was carried out using human liver microsomes and cDNA-expressed human CYPs (Supersomes). Perazine 65-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 15056479-1 2004 Identification of cytochrome P-450 isoenzymes (CYPs) involved in perazine 5-sulphoxidation and N-demethylation was carried out using human liver microsomes and cDNA-expressed human CYPs (Supersomes). Nitrogen 95-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 15370958-0 2004 Identification and relative contributions of human cytochrome P450 isoforms involved in the metabolism of glibenclamide and lansoprazole: evaluation of an approach based on the in vitro substrate disappearance rate. Glyburide 106-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-66 15370958-0 2004 Identification and relative contributions of human cytochrome P450 isoforms involved in the metabolism of glibenclamide and lansoprazole: evaluation of an approach based on the in vitro substrate disappearance rate. Lansoprazole 124-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-66 15370958-2 2004 The identification and relative contributions of human cytochrome P450 (CYP) enzymes involved in the metabolism of glibenclamide and lansoprazole in human liver microsomes were investigated using an approach based on the in vitro disappearance rate of unchanged drug. Glyburide 115-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 15370958-2 2004 The identification and relative contributions of human cytochrome P450 (CYP) enzymes involved in the metabolism of glibenclamide and lansoprazole in human liver microsomes were investigated using an approach based on the in vitro disappearance rate of unchanged drug. Glyburide 115-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 15370958-2 2004 The identification and relative contributions of human cytochrome P450 (CYP) enzymes involved in the metabolism of glibenclamide and lansoprazole in human liver microsomes were investigated using an approach based on the in vitro disappearance rate of unchanged drug. Lansoprazole 133-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 15370958-2 2004 The identification and relative contributions of human cytochrome P450 (CYP) enzymes involved in the metabolism of glibenclamide and lansoprazole in human liver microsomes were investigated using an approach based on the in vitro disappearance rate of unchanged drug. Lansoprazole 133-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 15135080-5 2004 Hydroxylation by cytochrome P450 (CYP) enzymes and conjugation with glucuronide and sulfate are among the major hepatic pathways of steroid inactivation. Glucuronides 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-32 15135080-5 2004 Hydroxylation by cytochrome P450 (CYP) enzymes and conjugation with glucuronide and sulfate are among the major hepatic pathways of steroid inactivation. Glucuronides 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 15135080-5 2004 Hydroxylation by cytochrome P450 (CYP) enzymes and conjugation with glucuronide and sulfate are among the major hepatic pathways of steroid inactivation. Sulfates 84-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-32 15135080-5 2004 Hydroxylation by cytochrome P450 (CYP) enzymes and conjugation with glucuronide and sulfate are among the major hepatic pathways of steroid inactivation. Sulfates 84-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 15135080-5 2004 Hydroxylation by cytochrome P450 (CYP) enzymes and conjugation with glucuronide and sulfate are among the major hepatic pathways of steroid inactivation. Steroids 132-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-32 15135080-5 2004 Hydroxylation by cytochrome P450 (CYP) enzymes and conjugation with glucuronide and sulfate are among the major hepatic pathways of steroid inactivation. Steroids 132-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 15135080-7 2004 The barbiturate phenobarbital and the environmental toxicant 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (DDE) are among the well characterized inducers for the CYP 2B and 3A enzymes and selected conjugation enzymes. barbituric acid 4-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 15135080-7 2004 The barbiturate phenobarbital and the environmental toxicant 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (DDE) are among the well characterized inducers for the CYP 2B and 3A enzymes and selected conjugation enzymes. Phenobarbital 16-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 15135080-7 2004 The barbiturate phenobarbital and the environmental toxicant 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (DDE) are among the well characterized inducers for the CYP 2B and 3A enzymes and selected conjugation enzymes. Dichlorodiphenyl Dichloroethylene 61-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 15135080-7 2004 The barbiturate phenobarbital and the environmental toxicant 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene (DDE) are among the well characterized inducers for the CYP 2B and 3A enzymes and selected conjugation enzymes. Dichlorodiphenyl Dichloroethylene 107-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 15078344-1 2004 It has been shown that large doses of acetaminophen can result in increased degradation of the hepatic cytochrome P450 (CYP) enzymes in vivo; however, the proteolytic pathways have not been identified. Acetaminophen 38-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 15051744-12 2004 Since this isozyme had the lowest in vitro IC(50) values for the cytochrome P450s most commonly involved with the metabolism of drugs (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4), azimilide-related drug interactions mediated via these isozymes are not anticipated. azimilide 181-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 15089095-1 2004 Cytochrome p450 (p450) 1A2 and NADPH-P450 reductase (NPR) catalyzed the oxidation of 2-amino-3-methylimidazo[4,5-f]quinoline (IQ), with consumption of NADPH. 2-amino-3-methylimidazo(4,5-f)quinoline 85-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 15066665-0 2004 Effect of posaconazole on cytochrome P450 enzymes: a randomized, open-label, two-way crossover study. posaconazole 10-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 15066665-2 2004 In this randomised, open-label, two-way crossover study, the potential for drug interactions with posaconazole via the cytochrome P450 (CYP450) enzyme pathway was evaluated. posaconazole 98-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-134 15066665-2 2004 In this randomised, open-label, two-way crossover study, the potential for drug interactions with posaconazole via the cytochrome P450 (CYP450) enzyme pathway was evaluated. posaconazole 98-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-142 15066665-9 2004 Drug interactions mediated by various CYP450 are common with the currently available triazole antifungals, however these results suggest that posaconazole may have an improved and more narrow drug interaction profile (CYP3A4 only) compared with other triazoles. posaconazole 142-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-44 15268978-0 2004 Role of individual human cytochrome P450 enzymes in the in vitro metabolism of hydromorphone. Hydromorphone 79-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 15102545-1 2004 The human cytochrome P450 (CYP) complement of heme-thiolate enzymes is reviewed. Heme 46-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 15102545-1 2004 The human cytochrome P450 (CYP) complement of heme-thiolate enzymes is reviewed. Heme 46-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 15041065-8 2004 This mini-review gives a summary and commentary on early studies on effects of lead on haem metabolism that provide us the clue to why investigations of the impacts of other toxic heavy metals and metalloids such as cadmium and arsenic on different human cytochrome P450 forms have become of great interest at the current time. Cadmium 216-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 255-270 15268978-2 2004 The aim was to identify the individual human cytochrome P450 (CYP) enzymes responsible for the in vitro N-demethylation of hydromorphone and to determine the potential effect of the inhibition of this metabolic pathway on the formation of other hydromorphone metabolites. Nitrogen 104-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 15268978-2 2004 The aim was to identify the individual human cytochrome P450 (CYP) enzymes responsible for the in vitro N-demethylation of hydromorphone and to determine the potential effect of the inhibition of this metabolic pathway on the formation of other hydromorphone metabolites. Nitrogen 104-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 15268978-2 2004 The aim was to identify the individual human cytochrome P450 (CYP) enzymes responsible for the in vitro N-demethylation of hydromorphone and to determine the potential effect of the inhibition of this metabolic pathway on the formation of other hydromorphone metabolites. Hydromorphone 123-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 15268978-2 2004 The aim was to identify the individual human cytochrome P450 (CYP) enzymes responsible for the in vitro N-demethylation of hydromorphone and to determine the potential effect of the inhibition of this metabolic pathway on the formation of other hydromorphone metabolites. Hydromorphone 123-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 15268978-2 2004 The aim was to identify the individual human cytochrome P450 (CYP) enzymes responsible for the in vitro N-demethylation of hydromorphone and to determine the potential effect of the inhibition of this metabolic pathway on the formation of other hydromorphone metabolites. Hydromorphone 245-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 15268978-2 2004 The aim was to identify the individual human cytochrome P450 (CYP) enzymes responsible for the in vitro N-demethylation of hydromorphone and to determine the potential effect of the inhibition of this metabolic pathway on the formation of other hydromorphone metabolites. Hydromorphone 245-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 15104117-0 2004 Metabolism of ethylbenzene by human liver microsomes and recombinant human cytochrome P450s (CYP). ethylbenzene 14-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 14993813-0 2004 Effect of nilvadipine, a dihydropyridine calcium antagonist, on cytochrome P450 activities in human hepatic microsomes. nilvadipine 10-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 14993813-1 2004 The effects of nilvadipine, a dihydropyridine calcium antagonist, on cytochrome P450 (CYP) activities in human hepatic microsomes were investigated. nilvadipine 15-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 14993813-1 2004 The effects of nilvadipine, a dihydropyridine calcium antagonist, on cytochrome P450 (CYP) activities in human hepatic microsomes were investigated. nilvadipine 15-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 14504090-6 2004 These data suggest that azole antifungals, through differential inhibition of hepatic cytochrome P-450 isoenzymes, affect CY metabolism and conditioning-related toxicities. Azoles 24-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 14998432-1 2004 AIMS: To identify the cytochrome p450 (CYP) enzyme(s) that catalyze the metabolism of tizanidine in vitro. tizanidine 86-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 14998432-1 2004 AIMS: To identify the cytochrome p450 (CYP) enzyme(s) that catalyze the metabolism of tizanidine in vitro. tizanidine 86-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 14998432-2 2004 METHODS: The effect of CYP isoform inhibitors on the elimination of tizanidine was studied using pooled human liver microsomes. tizanidine 68-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 14660610-0 2004 CYP2U1, a novel human thymus- and brain-specific cytochrome P450, catalyzes omega- and (omega-1)-hydroxylation of fatty acids. Fatty Acids 114-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-95 15202615-1 2004 PURPOSE OF REVIEW: Cytochrome P-450 metabolites of arachidonic acid have been reported to play an important role in the control of renal function and vascular tone, and in the long-term control of arterial pressure. Arachidonic Acid 51-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 15202615-4 2004 RECENT FINDINGS: Studies have demonstrated that the expression of cytochrome P-450 enzymes and the synthesis of 20-hydroxyeicosatetraenoic acid and epoxyeicosatrienoic acids in the kidney and peripheral vasculature are altered in many genetic and experimental models of hypertension. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 112-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 15202615-4 2004 RECENT FINDINGS: Studies have demonstrated that the expression of cytochrome P-450 enzymes and the synthesis of 20-hydroxyeicosatetraenoic acid and epoxyeicosatrienoic acids in the kidney and peripheral vasculature are altered in many genetic and experimental models of hypertension. epoxyeicosatrienoic acids 148-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 15202615-7 2004 SUMMARY: This review summarizes recent findings that address the issue of whether cytochrome P-450 metabolites of arachidonic acid play an important role in the regulation of renal tubular and peripheral vascular function and contribute to the pathogenesis of hypertension. Arachidonic Acid 114-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 15038401-3 2004 Potent inductors of CYP isoenzymes are carbamazepine, phenobarbital, phenytoin, and primidone, thereby decreasing not only their own plasma levels and efficacy but also that of other antiepileptics and other drugs. Carbamazepine 39-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-23 15038401-3 2004 Potent inductors of CYP isoenzymes are carbamazepine, phenobarbital, phenytoin, and primidone, thereby decreasing not only their own plasma levels and efficacy but also that of other antiepileptics and other drugs. Phenobarbital 54-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-23 15038401-3 2004 Potent inductors of CYP isoenzymes are carbamazepine, phenobarbital, phenytoin, and primidone, thereby decreasing not only their own plasma levels and efficacy but also that of other antiepileptics and other drugs. Phenytoin 69-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-23 15038401-3 2004 Potent inductors of CYP isoenzymes are carbamazepine, phenobarbital, phenytoin, and primidone, thereby decreasing not only their own plasma levels and efficacy but also that of other antiepileptics and other drugs. Primidone 84-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-23 15038401-4 2004 Felbamate, oxcarbazepine, and topiramate are weak inductors of the CYP isoenzyme 3A4, whereas they inhibit CYP2C19. Felbamate 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 15038401-4 2004 Felbamate, oxcarbazepine, and topiramate are weak inductors of the CYP isoenzyme 3A4, whereas they inhibit CYP2C19. Oxcarbazepine 11-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 15038401-4 2004 Felbamate, oxcarbazepine, and topiramate are weak inductors of the CYP isoenzyme 3A4, whereas they inhibit CYP2C19. Topiramate 30-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 15038401-5 2004 Valproic acid is a potent inhibitor of several CYP isoenzymes and glucuronyltransferases, resulting in an increase in plasma concentrations and toxicity of antiepileptics and other drugs. Valproic Acid 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 15013014-0 2004 Cytochrome P450/NADPH-dependent formation of trans epoxides from trans-arachidonic acids. trans epoxides 45-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-21 15013014-0 2004 Cytochrome P450/NADPH-dependent formation of trans epoxides from trans-arachidonic acids. trans-arachidonic acids 65-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-21 15013014-2 2004 We found that hepatic microsomes oxidized trans-AA via cytochrome P450/NADPH system to epoxides, which were hydrolyzed by epoxide hydrolase to diols (DiHETEs). Epoxy Compounds 87-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-76 15013014-2 2004 We found that hepatic microsomes oxidized trans-AA via cytochrome P450/NADPH system to epoxides, which were hydrolyzed by epoxide hydrolase to diols (DiHETEs). diols 143-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-76 15013014-2 2004 We found that hepatic microsomes oxidized trans-AA via cytochrome P450/NADPH system to epoxides, which were hydrolyzed by epoxide hydrolase to diols (DiHETEs). dihetes 150-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-76 14729370-2 2004 To investigate the cytochrome P450 (CYP) isoform involved in the activation of 10-azaBaP to the genotoxic form, the mutagenicity of 10-azaBaP using nine individual donors" and pooled human liver microsome preparations was compared with each CYP activity. 10-azabenzo(a)pyrene 79-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 14729370-2 2004 To investigate the cytochrome P450 (CYP) isoform involved in the activation of 10-azaBaP to the genotoxic form, the mutagenicity of 10-azaBaP using nine individual donors" and pooled human liver microsome preparations was compared with each CYP activity. 10-azabenzo(a)pyrene 79-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 14729370-2 2004 To investigate the cytochrome P450 (CYP) isoform involved in the activation of 10-azaBaP to the genotoxic form, the mutagenicity of 10-azaBaP using nine individual donors" and pooled human liver microsome preparations was compared with each CYP activity. 10-azabenzo(a)pyrene 132-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 14729370-7 2004 With regard to the proposal that BaP may be activated by human CYP1A1, our results suggest that the nitrogen-substitution at position-10 of BaP may cause the CYP enzyme-specificity in metabolic activation to change from CYP1A1 to CYP1A2. Benzo(a)pyrene 33-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 14729370-7 2004 With regard to the proposal that BaP may be activated by human CYP1A1, our results suggest that the nitrogen-substitution at position-10 of BaP may cause the CYP enzyme-specificity in metabolic activation to change from CYP1A1 to CYP1A2. Nitrogen 100-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 14729370-7 2004 With regard to the proposal that BaP may be activated by human CYP1A1, our results suggest that the nitrogen-substitution at position-10 of BaP may cause the CYP enzyme-specificity in metabolic activation to change from CYP1A1 to CYP1A2. Benzo(a)pyrene 140-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 14744142-1 2004 Cytochrome P450 (P450) 1A2 is the major enzyme involved in the metabolism of 2-amino-3,5-dimethylimidazo[4,5-f]quinoline (MeIQ) and other heterocyclic arylamines and their bioactivation to mutagens. 3H-Imidazo(4,5-f)quinolin-2-amine, 3,5-dimethyl- 77-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 14744142-1 2004 Cytochrome P450 (P450) 1A2 is the major enzyme involved in the metabolism of 2-amino-3,5-dimethylimidazo[4,5-f]quinoline (MeIQ) and other heterocyclic arylamines and their bioactivation to mutagens. 2-amino-3,4-dimethylimidazo(4,5-f)quinoline 122-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 14744142-1 2004 Cytochrome P450 (P450) 1A2 is the major enzyme involved in the metabolism of 2-amino-3,5-dimethylimidazo[4,5-f]quinoline (MeIQ) and other heterocyclic arylamines and their bioactivation to mutagens. aniline 151-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 15022728-2 2004 This study was performed to identify the metabolic pathway of KR-31543 in human liver microsomes and to characterize cytochrome P450 (CYP) enzymes that are involved in the metabolism of KR-31543. Krypton 62-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-132 15022728-2 2004 This study was performed to identify the metabolic pathway of KR-31543 in human liver microsomes and to characterize cytochrome P450 (CYP) enzymes that are involved in the metabolism of KR-31543. Krypton 62-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-137 14748817-1 2004 AIMS: Previous work has shown that rifampicin, a potent inducer of several cytochrome P450 (CYP) enzymes and transporters, decreased the plasma concentrations of simvastatin acid by more than 90%. Rifampin 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 14748817-1 2004 AIMS: Previous work has shown that rifampicin, a potent inducer of several cytochrome P450 (CYP) enzymes and transporters, decreased the plasma concentrations of simvastatin acid by more than 90%. Rifampin 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 14748817-1 2004 AIMS: Previous work has shown that rifampicin, a potent inducer of several cytochrome P450 (CYP) enzymes and transporters, decreased the plasma concentrations of simvastatin acid by more than 90%. simvastatin acid 162-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 14748817-1 2004 AIMS: Previous work has shown that rifampicin, a potent inducer of several cytochrome P450 (CYP) enzymes and transporters, decreased the plasma concentrations of simvastatin acid by more than 90%. simvastatin acid 162-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 14985146-0 2004 In vivo metabolism of the new designer drug 1-(4-methoxyphenyl)piperazine (MeOPP) in rat and identification of the human cytochrome P450 enzymes responsible for the major metabolic step. 1-(4-methoxyphenyl)piperazine 44-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-136 15049511-0 2004 Inhibitory effects of the monoamine oxidase inhibitor tranylcypromine on the cytochrome P450 enzymes CYP2C19, CYP2C9, and CYP2D6. Tranylcypromine 54-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 15049511-2 2004 The inhibitory effects of tranylcypromine, a nonselective irreversible inhibitor of monoamine oxidase (MAO), on three cytochrome P450 (CYP) enzymes, namely CYP2C9, CYP2C19, and CYP2D6, have been evaluated in vitro. Tranylcypromine 26-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 15049511-2 2004 The inhibitory effects of tranylcypromine, a nonselective irreversible inhibitor of monoamine oxidase (MAO), on three cytochrome P450 (CYP) enzymes, namely CYP2C9, CYP2C19, and CYP2D6, have been evaluated in vitro. Tranylcypromine 26-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 15049511-6 2004 A range of substrate concentrations was coincubated with a range of tranylcypromine concentrations in the presence of each of the CYP enzymes at 37 degrees C for a predetermined period of time. Tranylcypromine 68-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 14730412-1 2004 BACKGROUND: In vitro findings have indicated that the novel anxiolytic drug, deramciclane, is an inhibitor of the cytochrome P(450) (CYP) 2D6 enzyme and co-administration of deramciclane and the CYP2D6 probe drug desipramine is possible in clinical practice. Desipramine 213-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-131 14730412-1 2004 BACKGROUND: In vitro findings have indicated that the novel anxiolytic drug, deramciclane, is an inhibitor of the cytochrome P(450) (CYP) 2D6 enzyme and co-administration of deramciclane and the CYP2D6 probe drug desipramine is possible in clinical practice. Desipramine 213-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 14610241-1 2004 In the studies reported here, the ability of atomoxetine hydrochloride (Strattera) to inhibit or induce the metabolic capabilities of selected human isoforms of cytochrome P450 was evaluated. Atomoxetine Hydrochloride 45-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-176 14630070-5 2004 Testosterone was used as a marker substrate for cytochrome P450 mediated drug metabolism (phase I). Testosterone 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 14985146-0 2004 In vivo metabolism of the new designer drug 1-(4-methoxyphenyl)piperazine (MeOPP) in rat and identification of the human cytochrome P450 enzymes responsible for the major metabolic step. 1-(4-methoxyphenyl)piperazine 75-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-136 14698415-1 2004 Heterocyclic amines (HCAs) produced by cooking meat products at high temperatures are promutagens that are activated by cytochrome P450 (CYP) lA2. heterocyclic amines 0-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 14698415-1 2004 Heterocyclic amines (HCAs) produced by cooking meat products at high temperatures are promutagens that are activated by cytochrome P450 (CYP) lA2. heterocyclic amines 0-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 14698415-1 2004 Heterocyclic amines (HCAs) produced by cooking meat products at high temperatures are promutagens that are activated by cytochrome P450 (CYP) lA2. hcas 21-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 14698415-1 2004 Heterocyclic amines (HCAs) produced by cooking meat products at high temperatures are promutagens that are activated by cytochrome P450 (CYP) lA2. hcas 21-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 14709907-0 2004 Effect of cefixime and cefdinir, oral cephalosporins, on cytochrome P450 activities in human hepatic microsomes. Cefixime 10-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 15344842-2 2004 Almost the entire radioactivity was from the unchanged substance or lactone form in human hepatocytes, and the cytochrome P450 (CYP)-mediated metabolism of pitavastatin was negligible. pitavastatin 156-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-126 15344842-2 2004 Almost the entire radioactivity was from the unchanged substance or lactone form in human hepatocytes, and the cytochrome P450 (CYP)-mediated metabolism of pitavastatin was negligible. pitavastatin 156-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 15285699-6 2004 Apart from pravastatin and rosuvastatin, HMG-CoA reductase inhibitors are metabolized by the phase I cytochrome P450 (CYP) superfamily of drug metabolizing enzymes. Pravastatin 11-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 15285699-6 2004 Apart from pravastatin and rosuvastatin, HMG-CoA reductase inhibitors are metabolized by the phase I cytochrome P450 (CYP) superfamily of drug metabolizing enzymes. Pravastatin 11-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 15285699-6 2004 Apart from pravastatin and rosuvastatin, HMG-CoA reductase inhibitors are metabolized by the phase I cytochrome P450 (CYP) superfamily of drug metabolizing enzymes. Rosuvastatin Calcium 27-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 15285699-6 2004 Apart from pravastatin and rosuvastatin, HMG-CoA reductase inhibitors are metabolized by the phase I cytochrome P450 (CYP) superfamily of drug metabolizing enzymes. Rosuvastatin Calcium 27-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 14709907-0 2004 Effect of cefixime and cefdinir, oral cephalosporins, on cytochrome P450 activities in human hepatic microsomes. Cefdinir 23-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 14709907-0 2004 Effect of cefixime and cefdinir, oral cephalosporins, on cytochrome P450 activities in human hepatic microsomes. Cephalosporins 38-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 14709907-1 2004 The effects of two kinds of oral cephalosporins, cefixime and cefdinir, on cytochrome P450 (CYP) activities in human hepatic microsomes were investigated. Cephalosporins 33-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 14709907-1 2004 The effects of two kinds of oral cephalosporins, cefixime and cefdinir, on cytochrome P450 (CYP) activities in human hepatic microsomes were investigated. Cephalosporins 33-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 14709907-1 2004 The effects of two kinds of oral cephalosporins, cefixime and cefdinir, on cytochrome P450 (CYP) activities in human hepatic microsomes were investigated. Cefixime 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 14709907-1 2004 The effects of two kinds of oral cephalosporins, cefixime and cefdinir, on cytochrome P450 (CYP) activities in human hepatic microsomes were investigated. Cefixime 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 14709907-1 2004 The effects of two kinds of oral cephalosporins, cefixime and cefdinir, on cytochrome P450 (CYP) activities in human hepatic microsomes were investigated. Cefdinir 62-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 14709907-1 2004 The effects of two kinds of oral cephalosporins, cefixime and cefdinir, on cytochrome P450 (CYP) activities in human hepatic microsomes were investigated. Cefdinir 62-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 14729637-1 2004 Cytochrome P450 (CYP) gene transfer sensitizes tumor xenografts to anticancer prodrugs such as cyclophosphamide (CPA) without a detectable increase in host toxicity. Cyclophosphamide 95-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 14729637-1 2004 Cytochrome P450 (CYP) gene transfer sensitizes tumor xenografts to anticancer prodrugs such as cyclophosphamide (CPA) without a detectable increase in host toxicity. Cyclophosphamide 95-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 14729637-1 2004 Cytochrome P450 (CYP) gene transfer sensitizes tumor xenografts to anticancer prodrugs such as cyclophosphamide (CPA) without a detectable increase in host toxicity. Cyclophosphamide 113-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 14729637-1 2004 Cytochrome P450 (CYP) gene transfer sensitizes tumor xenografts to anticancer prodrugs such as cyclophosphamide (CPA) without a detectable increase in host toxicity. Cyclophosphamide 113-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 14628297-1 2004 Methadone is a clinically used opioid agonist that is oxidatively metabolized by cytochrome P450 (CYP) isoforms to a stable metabolite, EDDP. Methadone 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 31394618-0 2004 Altered Levels of Cytochrome P450 Genes in Hepatitis B or C Virus-infected Liver Identified by Oligonucleotide Microarray. Oligonucleotides 95-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 14628297-1 2004 Methadone is a clinically used opioid agonist that is oxidatively metabolized by cytochrome P450 (CYP) isoforms to a stable metabolite, EDDP. Methadone 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 14628297-4 2004 The ability of the common human drug-metabolizing CYPs (obtained from baculovirus-infected insect cell supersomes) to generate 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrilidine (EDDP) from racemic methadone was examined and then determined if the CYP isoforms metabolized methadone stereoselectively. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrilidine 127-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 14628297-1 2004 Methadone is a clinically used opioid agonist that is oxidatively metabolized by cytochrome P450 (CYP) isoforms to a stable metabolite, EDDP. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 136-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 14628297-4 2004 The ability of the common human drug-metabolizing CYPs (obtained from baculovirus-infected insect cell supersomes) to generate 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrilidine (EDDP) from racemic methadone was examined and then determined if the CYP isoforms metabolized methadone stereoselectively. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 178-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 14628297-1 2004 Methadone is a clinically used opioid agonist that is oxidatively metabolized by cytochrome P450 (CYP) isoforms to a stable metabolite, EDDP. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 136-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 14628297-4 2004 The ability of the common human drug-metabolizing CYPs (obtained from baculovirus-infected insect cell supersomes) to generate 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrilidine (EDDP) from racemic methadone was examined and then determined if the CYP isoforms metabolized methadone stereoselectively. Methadone 197-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 14628297-4 2004 The ability of the common human drug-metabolizing CYPs (obtained from baculovirus-infected insect cell supersomes) to generate 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrilidine (EDDP) from racemic methadone was examined and then determined if the CYP isoforms metabolized methadone stereoselectively. Methadone 272-281 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 14628297-3 2004 The cytochrome P450 (CYP) isoform involved in methadone"s metabolism is thought to be CYP3A4, but human drug-drug interaction studies are not consistent with this. Methadone 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 14628297-3 2004 The cytochrome P450 (CYP) isoform involved in methadone"s metabolism is thought to be CYP3A4, but human drug-drug interaction studies are not consistent with this. Methadone 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 14628297-8 2004 Michaelis-Menten kinetic data demonstrated that CYP2B6 had the highest V(max) (44 ng/min/10pmol) and the lowest K(m) (12.6 microg/ml) for EDDP formation of all the CYP isoforms. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 138-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 14715049-15 2004 The main CYP isoforms involved are CYP3A4 for lidocaine and bupivacaine and CYP1A2 for ropivacaine. Ropivacaine 87-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 15509184-15 2004 Although bicalutamide is a CYP inducer in laboratory animals, dosages < or = 150 mg/day have shown no evidence of enzyme induction in humans. bicalutamide 9-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 14715049-15 2004 The main CYP isoforms involved are CYP3A4 for lidocaine and bupivacaine and CYP1A2 for ropivacaine. Lidocaine 46-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 14715049-15 2004 The main CYP isoforms involved are CYP3A4 for lidocaine and bupivacaine and CYP1A2 for ropivacaine. Bupivacaine 60-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 15244495-7 2004 Factors reported to influence the pharmacokinetics of tacrolimus include the patient group studied, hepatic dysfunction, hepatitis C status, time after transplantation, patient age, donor liver characteristics, recipient race, haematocrit and albumin concentrations, diurnal rhythm, food administration, corticosteroid dosage, diarrhoea and cytochrome P450 (CYP) isoenzyme and P-glycoprotein expression. Tacrolimus 54-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 341-356 15244495-7 2004 Factors reported to influence the pharmacokinetics of tacrolimus include the patient group studied, hepatic dysfunction, hepatitis C status, time after transplantation, patient age, donor liver characteristics, recipient race, haematocrit and albumin concentrations, diurnal rhythm, food administration, corticosteroid dosage, diarrhoea and cytochrome P450 (CYP) isoenzyme and P-glycoprotein expression. Tacrolimus 54-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 358-361 15509184-11 2004 Bicalutamide appears to be cleared almost exclusively by metabolism; this is largely mediated by cytochrome P450 (CYP) for (R)-bicalutamide, but glucuronidation is the predominant metabolic route for (S)-bicalutamide. bicalutamide 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-112 15509184-11 2004 Bicalutamide appears to be cleared almost exclusively by metabolism; this is largely mediated by cytochrome P450 (CYP) for (R)-bicalutamide, but glucuronidation is the predominant metabolic route for (S)-bicalutamide. bicalutamide 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 15509184-11 2004 Bicalutamide appears to be cleared almost exclusively by metabolism; this is largely mediated by cytochrome P450 (CYP) for (R)-bicalutamide, but glucuronidation is the predominant metabolic route for (S)-bicalutamide. (R)-Bicalutamide 123-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-112 15611610-4 2004 Biochemical studies indicated that the Erg11p from the TIMM3209 strain showed reduced susceptibility both to fluconazole and itraconazole of cell-free ergosterol biosynthesis, and cytochrome P-450 also showed reduced affinity to fluconazole in the carbon monoxidecytochrome P-450 complex formation assay. Fluconazole 229-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 15509184-11 2004 Bicalutamide appears to be cleared almost exclusively by metabolism; this is largely mediated by cytochrome P450 (CYP) for (R)-bicalutamide, but glucuronidation is the predominant metabolic route for (S)-bicalutamide. (R)-Bicalutamide 123-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 15760805-4 2004 The pharmacokinetics of oral oxycodone differs from oral morphine in that it has a higher bioavailability, a slightly longer half-life, and is hepatically metabolized by cytochrome P450 rather than undergoing glucuronidation. Oxycodone 29-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 170-185 15611610-6 2004 We found that the cytochrome P-450 from the TIMM3209 strain decayed during incubation at 37 C without fluconazole although it is unknown whether or not the phenomenon is linked to the resistant phenotype. Fluconazole 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 14966856-4 2004 When ethoxidine was incubated with BNF-treated rat liver microsomes or with cells expressing different recombinant human cytochrome P450, the same four ethoxidine metabolites (m(1)-m(4)) were detected and were formed exclusively by CYP1A1. ethoxidine 5-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-136 15379059-1 2004 Cytochrome P450 (CYP) enzymes participate in the metabolism of a variety of naturally occurring and foreign compounds by reactions requiring NADPH and O2. NADP 141-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15379059-1 2004 Cytochrome P450 (CYP) enzymes participate in the metabolism of a variety of naturally occurring and foreign compounds by reactions requiring NADPH and O2. NADP 141-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 15379059-1 2004 Cytochrome P450 (CYP) enzymes participate in the metabolism of a variety of naturally occurring and foreign compounds by reactions requiring NADPH and O2. Oxygen 151-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 15379059-1 2004 Cytochrome P450 (CYP) enzymes participate in the metabolism of a variety of naturally occurring and foreign compounds by reactions requiring NADPH and O2. Oxygen 151-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 14966856-4 2004 When ethoxidine was incubated with BNF-treated rat liver microsomes or with cells expressing different recombinant human cytochrome P450, the same four ethoxidine metabolites (m(1)-m(4)) were detected and were formed exclusively by CYP1A1. ethoxidine 152-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-136 14580893-1 2003 Fluconazole (FCZ) is a potent inhibitor of the cytochrome P450 (CYP)-mediated metabolism of the anti-epileptic agent phenytoin (PHT), a well-known human and animal teratogen. Fluconazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 12966104-4 2003 These are reminiscent of the mechanism of cytochrome P-450, where a heme iron stabilizes the activated O2 species. Heme 68-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 12966104-4 2003 These are reminiscent of the mechanism of cytochrome P-450, where a heme iron stabilizes the activated O2 species. Iron 73-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 12966104-4 2003 These are reminiscent of the mechanism of cytochrome P-450, where a heme iron stabilizes the activated O2 species. Oxygen 103-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 12966104-9 2003 In the context of the open and fully solvent-accessible active site for the homologous peptidylglycine-alpha-hydroxylating monooxygenase and by analogy to cytochrome P-450, the accumulation of a reduced and activated oxygen species in DbetaM before C-H cleavage would be expected to give some uncoupling of oxygen and substrate consumption. Oxygen 127-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 12966104-9 2003 In the context of the open and fully solvent-accessible active site for the homologous peptidylglycine-alpha-hydroxylating monooxygenase and by analogy to cytochrome P-450, the accumulation of a reduced and activated oxygen species in DbetaM before C-H cleavage would be expected to give some uncoupling of oxygen and substrate consumption. Oxygen 217-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 14580893-1 2003 Fluconazole (FCZ) is a potent inhibitor of the cytochrome P450 (CYP)-mediated metabolism of the anti-epileptic agent phenytoin (PHT), a well-known human and animal teratogen. Fluconazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 14580893-1 2003 Fluconazole (FCZ) is a potent inhibitor of the cytochrome P450 (CYP)-mediated metabolism of the anti-epileptic agent phenytoin (PHT), a well-known human and animal teratogen. Fluconazole 13-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 14580893-1 2003 Fluconazole (FCZ) is a potent inhibitor of the cytochrome P450 (CYP)-mediated metabolism of the anti-epileptic agent phenytoin (PHT), a well-known human and animal teratogen. Fluconazole 13-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 14580893-1 2003 Fluconazole (FCZ) is a potent inhibitor of the cytochrome P450 (CYP)-mediated metabolism of the anti-epileptic agent phenytoin (PHT), a well-known human and animal teratogen. Phenytoin 117-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 14580893-1 2003 Fluconazole (FCZ) is a potent inhibitor of the cytochrome P450 (CYP)-mediated metabolism of the anti-epileptic agent phenytoin (PHT), a well-known human and animal teratogen. Phenytoin 117-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 14580893-1 2003 Fluconazole (FCZ) is a potent inhibitor of the cytochrome P450 (CYP)-mediated metabolism of the anti-epileptic agent phenytoin (PHT), a well-known human and animal teratogen. Phenytoin 128-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 14580893-1 2003 Fluconazole (FCZ) is a potent inhibitor of the cytochrome P450 (CYP)-mediated metabolism of the anti-epileptic agent phenytoin (PHT), a well-known human and animal teratogen. Phenytoin 128-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 14580893-2 2003 It has been postulated that phenytoin must be bioactivated via the CYP system to initiate teratogenesis. Phenytoin 28-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 14684755-2 2003 In the liver, fatty acids can be metabolized by cytochrome P450 (CYP) enzymes, but little is known about the role of these enzymes in the vascular endothelium. Fatty Acids 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 14689466-5 2003 The chemical inhibition and correlation data indicated that the following substrates can be used as specific functional probes for individual cytochrome P450 present in human liver microsomes: coumarin for CYP2A6 (r=0.82), AMMC for CYP2D6 (r=0.83) and DBF for CYP3A4 (r=0.92). coumarin 193-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 14689466-5 2003 The chemical inhibition and correlation data indicated that the following substrates can be used as specific functional probes for individual cytochrome P450 present in human liver microsomes: coumarin for CYP2A6 (r=0.82), AMMC for CYP2D6 (r=0.83) and DBF for CYP3A4 (r=0.92). ammc 223-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 14689466-5 2003 The chemical inhibition and correlation data indicated that the following substrates can be used as specific functional probes for individual cytochrome P450 present in human liver microsomes: coumarin for CYP2A6 (r=0.82), AMMC for CYP2D6 (r=0.83) and DBF for CYP3A4 (r=0.92). dibenzylfluorescein 252-255 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 14652237-1 2003 BACKGROUND: Tamoxifen, a selective estrogen receptor modulator (SERM), is converted to 4-hydroxy-tamoxifen and other active metabolites by cytochrome P450 (CYP) enzymes. Tamoxifen 12-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 14652237-1 2003 BACKGROUND: Tamoxifen, a selective estrogen receptor modulator (SERM), is converted to 4-hydroxy-tamoxifen and other active metabolites by cytochrome P450 (CYP) enzymes. Tamoxifen 12-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-159 14652237-1 2003 BACKGROUND: Tamoxifen, a selective estrogen receptor modulator (SERM), is converted to 4-hydroxy-tamoxifen and other active metabolites by cytochrome P450 (CYP) enzymes. 4'-hydroxytamoxifen 87-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 14652237-1 2003 BACKGROUND: Tamoxifen, a selective estrogen receptor modulator (SERM), is converted to 4-hydroxy-tamoxifen and other active metabolites by cytochrome P450 (CYP) enzymes. 4'-hydroxytamoxifen 87-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-159 14652237-6 2003 To determine which CYP isoforms were involved in the metabolism of tamoxifen to specific metabolites, we used CYP isoform-specific inhibitors. Tamoxifen 67-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 14652237-6 2003 To determine which CYP isoforms were involved in the metabolism of tamoxifen to specific metabolites, we used CYP isoform-specific inhibitors. Tamoxifen 67-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 14684755-2 2003 In the liver, fatty acids can be metabolized by cytochrome P450 (CYP) enzymes, but little is known about the role of these enzymes in the vascular endothelium. Fatty Acids 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 14684755-4 2003 We investigated the role of CYP-mediated mechanisms of linoleic acid metabolism in endothelial cell activation by examining the effects of linoleic acid or its oxidized metabolites such as LTX and leukotoxin diol (LTD). Linoleic Acid 55-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 14668935-4 2003 In humans, however, azole antifungals also interfere with several hepatic and intestinal cytochrome P-450 isoenzymes responsible for the metabolism of numerous drugs. Azoles 20-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 14570773-5 2003 In contrast to microsomes, the oxidation of rofecoxib to 5-hydroxyrofecoxib by S9 fractions followed two pathways, one NADPH-dependent and one NAD+-dependent (non-cytochrome P450), with the latter accounting for about 40% of total activity. rofecoxib 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-178 14570773-5 2003 In contrast to microsomes, the oxidation of rofecoxib to 5-hydroxyrofecoxib by S9 fractions followed two pathways, one NADPH-dependent and one NAD+-dependent (non-cytochrome P450), with the latter accounting for about 40% of total activity. 5-hydroxyrofecoxib 57-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-178 12970580-6 2003 Co-treatment of PC3/AR cells with SKF-525A, a nonselective inhibitor of cytochrome P450 (CYP) enzymes, enhanced the antiandrogenic effect, indicating that the antiandrogenic effect is caused by intact species of DEPE constituents. Proadifen 34-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 12970580-6 2003 Co-treatment of PC3/AR cells with SKF-525A, a nonselective inhibitor of cytochrome P450 (CYP) enzymes, enhanced the antiandrogenic effect, indicating that the antiandrogenic effect is caused by intact species of DEPE constituents. Proadifen 34-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 14742142-0 2003 Identification of the human cytochrome P450s responsible for the in vitro metabolism of a leukotriene B4 receptor antagonist, CP-195,543. cp-195 126-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 14742142-2 2003 The major human cytochrome P450 (CYP) form(s) responsible for the metabolism of CP-195,543, a potent leukotriene B4 antagonist, were investigated. cp-195 80-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 14742142-2 2003 The major human cytochrome P450 (CYP) form(s) responsible for the metabolism of CP-195,543, a potent leukotriene B4 antagonist, were investigated. cp-195 80-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 14742142-2 2003 The major human cytochrome P450 (CYP) form(s) responsible for the metabolism of CP-195,543, a potent leukotriene B4 antagonist, were investigated. Leukotriene B4 101-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 14742142-2 2003 The major human cytochrome P450 (CYP) form(s) responsible for the metabolism of CP-195,543, a potent leukotriene B4 antagonist, were investigated. Leukotriene B4 101-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 14742144-2 2003 Studies using human liver microsomes and recombinant human cytochrome P450 (CYP) and flavin-containing monooxygenase (FMO) were performed to identify the enzymes responsible for the metabolism of S-methyl-esonarimod (M2), an active metabolite of esonarimod (KE-298, a novel antirheumatic drug). esonarimod 205-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 14742144-2 2003 Studies using human liver microsomes and recombinant human cytochrome P450 (CYP) and flavin-containing monooxygenase (FMO) were performed to identify the enzymes responsible for the metabolism of S-methyl-esonarimod (M2), an active metabolite of esonarimod (KE-298, a novel antirheumatic drug). esonarimod 205-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 14742144-2 2003 Studies using human liver microsomes and recombinant human cytochrome P450 (CYP) and flavin-containing monooxygenase (FMO) were performed to identify the enzymes responsible for the metabolism of S-methyl-esonarimod (M2), an active metabolite of esonarimod (KE-298, a novel antirheumatic drug). esonarimod 246-256 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 14742144-2 2003 Studies using human liver microsomes and recombinant human cytochrome P450 (CYP) and flavin-containing monooxygenase (FMO) were performed to identify the enzymes responsible for the metabolism of S-methyl-esonarimod (M2), an active metabolite of esonarimod (KE-298, a novel antirheumatic drug). esonarimod 258-264 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 14678248-0 2003 Oxidative metabolism of tamoxifen to Z-4-hydroxy-tamoxifen by cytochrome P450 isoforms: an appraisal of in vitro studies. Tamoxifen 24-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 14678248-0 2003 Oxidative metabolism of tamoxifen to Z-4-hydroxy-tamoxifen by cytochrome P450 isoforms: an appraisal of in vitro studies. z-4-hydroxy-tamoxifen 37-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 14678248-8 2003 Cytochrome P450 (CYP) isoform-specific chemical and monoclonal antibody inhibition studies have demonstrated that CYP2B6, CYP2C9, CYP2D6 and CYP3A4 all mediate the formation of Z-4-OH-tam. z-4-oh-tam 177-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 14678248-8 2003 Cytochrome P450 (CYP) isoform-specific chemical and monoclonal antibody inhibition studies have demonstrated that CYP2B6, CYP2C9, CYP2D6 and CYP3A4 all mediate the formation of Z-4-OH-tam. z-4-oh-tam 177-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 14678248-10 2003 Significant associations between the percentage inhibition of Z-4-OH-tam by CYP isoform-specific inhibitors and the rate of metabolism of CYP isoform-specific index reactions and between individual expression of CYP2B6, CYP2C9 and CYP2D6 and Z-4-OH-tam formation rates indicate predominant roles for these isoforms in this pathway. z-4-oh-tam 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 14678248-10 2003 Significant associations between the percentage inhibition of Z-4-OH-tam by CYP isoform-specific inhibitors and the rate of metabolism of CYP isoform-specific index reactions and between individual expression of CYP2B6, CYP2C9 and CYP2D6 and Z-4-OH-tam formation rates indicate predominant roles for these isoforms in this pathway. z-4-oh-tam 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 14678248-10 2003 Significant associations between the percentage inhibition of Z-4-OH-tam by CYP isoform-specific inhibitors and the rate of metabolism of CYP isoform-specific index reactions and between individual expression of CYP2B6, CYP2C9 and CYP2D6 and Z-4-OH-tam formation rates indicate predominant roles for these isoforms in this pathway. z-4-oh-tam 242-252 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 14678248-10 2003 Significant associations between the percentage inhibition of Z-4-OH-tam by CYP isoform-specific inhibitors and the rate of metabolism of CYP isoform-specific index reactions and between individual expression of CYP2B6, CYP2C9 and CYP2D6 and Z-4-OH-tam formation rates indicate predominant roles for these isoforms in this pathway. z-4-oh-tam 242-252 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 14517708-1 2003 RATIONALE AND OBJECTIVE: Bromazepam, an anti-anxiety agent, has been reported to be metabolized by cytochrome P(450) (CYP). Bromazepam 25-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-116 14517708-1 2003 RATIONALE AND OBJECTIVE: Bromazepam, an anti-anxiety agent, has been reported to be metabolized by cytochrome P(450) (CYP). Bromazepam 25-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 14550947-0 2003 DNA adduct formation by the environmental contaminant 3-nitrobenzanthrone in V79 cells expressing human cytochrome P450 enzymes. 3-nitrobenzanthrone 54-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 12892558-3 2003 In particular, there has been considerable support for a role for the cytochrome P450 metabolites, the epoxyeicosatrienoic acids (EETs). epoxyeicosatrienoic acids 103-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 12892558-3 2003 In particular, there has been considerable support for a role for the cytochrome P450 metabolites, the epoxyeicosatrienoic acids (EETs). eets 130-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 14522569-1 2003 AIMS: Clopidogrel is a pro-drug which is converted to an active, unstable drug by cytochrome P450 (CYP). Clopidogrel 6-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 14522569-1 2003 AIMS: Clopidogrel is a pro-drug which is converted to an active, unstable drug by cytochrome P450 (CYP). Clopidogrel 6-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-102 14522569-3 2003 It has been recently suggested that the most abundant human CYP isoform, 3A4, activates clopidogrel. Clopidogrel 88-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 12904931-0 2003 Inhibitory effect of 5-fluorouracil on human cytochrome P(450) isoforms in human liver microsomes. Fluorouracil 21-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-62 22900324-0 2003 Cytochrome P450-mediated oxidative damage of nuclear membrane proteins and its prevention by vitamin C. Ascorbic Acid 93-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12943422-0 2003 Products of cytochrome P450(BioI) (CYP107H1)-catalyzed oxidation of fatty acids. Fatty Acids 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-33 12943422-1 2003 [reaction: see text] Oxidation of tetradecanoic and hexadecanoic acids by cytochrome P450(BioI) (CYP107H1) produces mainly the 11-, 12-, and 13-hydroxy C(14) fatty acids and the 11- to 15-hydroxy C(16) fatty acids, respectively. tetradecanoic 34-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-95 12943422-1 2003 [reaction: see text] Oxidation of tetradecanoic and hexadecanoic acids by cytochrome P450(BioI) (CYP107H1) produces mainly the 11-, 12-, and 13-hydroxy C(14) fatty acids and the 11- to 15-hydroxy C(16) fatty acids, respectively. Palmitic Acid 52-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-95 12943422-1 2003 [reaction: see text] Oxidation of tetradecanoic and hexadecanoic acids by cytochrome P450(BioI) (CYP107H1) produces mainly the 11-, 12-, and 13-hydroxy C(14) fatty acids and the 11- to 15-hydroxy C(16) fatty acids, respectively. Fatty Acids 158-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-95 12943422-1 2003 [reaction: see text] Oxidation of tetradecanoic and hexadecanoic acids by cytochrome P450(BioI) (CYP107H1) produces mainly the 11-, 12-, and 13-hydroxy C(14) fatty acids and the 11- to 15-hydroxy C(16) fatty acids, respectively. Fatty Acids 202-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-95 12919179-1 2003 AIMS: To identify the principal human cytochrome P450 (CYP) enzyme(s) responsible for the human in vitro biotransformation of repaglinide. repaglinide 126-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 12919179-1 2003 AIMS: To identify the principal human cytochrome P450 (CYP) enzyme(s) responsible for the human in vitro biotransformation of repaglinide. repaglinide 126-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 12919179-3 2003 METHODS: [14C]-Repaglinide was incubated with recombinant CYP and with human liver microsomes (HLM) from individual donors in the presence of inhibitory antibodies specific for individual CYP enzymes. repaglinide 15-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 12919179-3 2003 METHODS: [14C]-Repaglinide was incubated with recombinant CYP and with human liver microsomes (HLM) from individual donors in the presence of inhibitory antibodies specific for individual CYP enzymes. repaglinide 15-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-191 12919179-18 2003 This dual CYP biotransformation may have consequences for the clinical pharmacokinetics and drug-drug interactions involving repaglinide if one CYP pathway has sufficient capacity to compensate if the other is inhibited. repaglinide 125-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-13 12919179-18 2003 This dual CYP biotransformation may have consequences for the clinical pharmacokinetics and drug-drug interactions involving repaglinide if one CYP pathway has sufficient capacity to compensate if the other is inhibited. repaglinide 125-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-147 14604738-9 2003 Because ezetimibe is eliminated primarily by glucuronidation and not by cytochrome P450 (CYP) oxidation, it is subject to minimal drug interactions involving the CYP enzyme system. Ezetimibe 8-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 12920490-9 2003 We identified hepatic CYP enzymes responsible for metabolism of some compounds (praziquantel-1A2, 2C19, 3A4; primaquine-1A2, 3A4; chloroquine-2C8, 2D6, 3A4; artesunate-2A6; pyrantel-2D6). pyrantel-2d6 173-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 14626496-0 2003 Role of cytochrome P450-dependent arachidonic acid metabolites in liver physiology and pathophysiology. Arachidonic Acid 34-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 14626496-1 2003 Arachidonic acid (AA) can undergo monooxygenation or epoxidation by enzymes in the cytochrome P450 (CYP) family in the brain, kidney, lung, vasculature, and the liver. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-98 14626496-1 2003 Arachidonic acid (AA) can undergo monooxygenation or epoxidation by enzymes in the cytochrome P450 (CYP) family in the brain, kidney, lung, vasculature, and the liver. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 14626496-2 2003 CYP-AA metabolites, 19- and 20-hydroxyeicosatetraenoic acids (HETEs), epoxyeicosatrienoic acids (EETs) and diHETEs have different biological properties based on sites of production and can be stored in tissue lipids and released in response to hormonal stimuli. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 62-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 14626496-2 2003 CYP-AA metabolites, 19- and 20-hydroxyeicosatetraenoic acids (HETEs), epoxyeicosatrienoic acids (EETs) and diHETEs have different biological properties based on sites of production and can be stored in tissue lipids and released in response to hormonal stimuli. epoxyeicosatrienoic acids 70-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 14626496-2 2003 CYP-AA metabolites, 19- and 20-hydroxyeicosatetraenoic acids (HETEs), epoxyeicosatrienoic acids (EETs) and diHETEs have different biological properties based on sites of production and can be stored in tissue lipids and released in response to hormonal stimuli. eets 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 14626496-2 2003 CYP-AA metabolites, 19- and 20-hydroxyeicosatetraenoic acids (HETEs), epoxyeicosatrienoic acids (EETs) and diHETEs have different biological properties based on sites of production and can be stored in tissue lipids and released in response to hormonal stimuli. dihetes 107-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 14626496-11 2003 Compared with other organs, the liver has the highest total CYP content and contains the highest levels of individual CYP enzymes involved in the metabolism of fatty acids. Fatty Acids 160-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 14626496-12 2003 In humans, 50-75% of CYP-dependent AA metabolites formed by liver microsomes are omega/omega-OH-AA, mainly w-OH-AA, i.e. 20HETE, and 13-28% are EETs. omega-oh-aa 87-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 14626496-12 2003 In humans, 50-75% of CYP-dependent AA metabolites formed by liver microsomes are omega/omega-OH-AA, mainly w-OH-AA, i.e. 20HETE, and 13-28% are EETs. w-oh-aa 107-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 14555336-0 2003 Identification of cytochrome P450 enzymes involved in the metabolism of 4"-methoxy-alpha-pyrrolidinopropiophenone (MOPPP), a designer drug, in human liver microsomes. 4'-methoxy-alpha-pyrrolidinopropiophenone 72-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 14555336-0 2003 Identification of cytochrome P450 enzymes involved in the metabolism of 4"-methoxy-alpha-pyrrolidinopropiophenone (MOPPP), a designer drug, in human liver microsomes. 4'-methoxy-alpha-pyrrolidinopropiophenone 115-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 14555336-2 2003 The metabolism of 4"-methoxy-alpha-pyrrolidinopropiophenone (MOPPP), a novel designer drug, to its demethylated major metabolite 4"-hydroxy-pyrrolidinopropio-phenone (HO-PPP) was studied in pooled human liver microsomes (HLM) and in cDNA-expressed human hepatic cytochrome P450 (CYP) enzymes. 4'-methoxy-alpha-pyrrolidinopropiophenone 61-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-277 14555336-2 2003 The metabolism of 4"-methoxy-alpha-pyrrolidinopropiophenone (MOPPP), a novel designer drug, to its demethylated major metabolite 4"-hydroxy-pyrrolidinopropio-phenone (HO-PPP) was studied in pooled human liver microsomes (HLM) and in cDNA-expressed human hepatic cytochrome P450 (CYP) enzymes. 4'-methoxy-alpha-pyrrolidinopropiophenone 61-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 279-282 12920490-9 2003 We identified hepatic CYP enzymes responsible for metabolism of some compounds (praziquantel-1A2, 2C19, 3A4; primaquine-1A2, 3A4; chloroquine-2C8, 2D6, 3A4; artesunate-2A6; pyrantel-2D6). artesunate-2a6 157-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 12904931-2 2003 However, little is known regarding the inhibitory potential of 5-FU on the metabolism of co-administered drugs by cytochrome P(450) (CYP). Fluorouracil 63-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-131 12904931-2 2003 However, little is known regarding the inhibitory potential of 5-FU on the metabolism of co-administered drugs by cytochrome P(450) (CYP). Fluorouracil 63-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 12904931-3 2003 The aim of the present study was to elucidate the inhibitory effect of 5-FU on CYP isoforms using human liver microsomes. Fluorouracil 71-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 12920490-9 2003 We identified hepatic CYP enzymes responsible for metabolism of some compounds (praziquantel-1A2, 2C19, 3A4; primaquine-1A2, 3A4; chloroquine-2C8, 2D6, 3A4; artesunate-2A6; pyrantel-2D6). praziquantel-1a2 80-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 12920490-9 2003 We identified hepatic CYP enzymes responsible for metabolism of some compounds (praziquantel-1A2, 2C19, 3A4; primaquine-1A2, 3A4; chloroquine-2C8, 2D6, 3A4; artesunate-2A6; pyrantel-2D6). primaquine-1a2 109-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 12920490-9 2003 We identified hepatic CYP enzymes responsible for metabolism of some compounds (praziquantel-1A2, 2C19, 3A4; primaquine-1A2, 3A4; chloroquine-2C8, 2D6, 3A4; artesunate-2A6; pyrantel-2D6). Chloroquine 130-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-25 12971034-8 2003 In vitro experiments suggested that karenitecin is metabolized by CYP450 3A4, 2C8, and 2D6 isoenzymes and is an inhibitor of the CYP450 3A4 and 2C8 isoenzymes. cositecan 36-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-135 12971034-12 2003 It was concluded that karenitecin has the potential to alter CYP450 3A4 and 2C8 drug-metabolizing activity. cositecan 22-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-67 12971034-2 2003 Individual cloned cytochrome P450 (CYP450) isoenzymes were used to determine, in vitro, the metabolism of karenitecin. cositecan 106-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Bupropion 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 12971034-2 2003 Individual cloned cytochrome P450 (CYP450) isoenzymes were used to determine, in vitro, the metabolism of karenitecin. cositecan 106-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-41 12971034-3 2003 Known substrates and inhibitors of each isoenzyme were employed to evaluate CYP450 drug interactions with karenitecin. cositecan 106-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-82 12971034-8 2003 In vitro experiments suggested that karenitecin is metabolized by CYP450 3A4, 2C8, and 2D6 isoenzymes and is an inhibitor of the CYP450 3A4 and 2C8 isoenzymes. cositecan 36-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-72 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Bupropion 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Bupropion 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-162 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Methanol 257-265 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Methanol 257-265 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Rifampin 276-286 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Rifampin 276-286 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Rifampin 288-296 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 14604466-2 2003 The possibility of cytochrome p450 (CYP) induction by bupropion (10 microM) was evaluated in-vitro by comparing catalytic activity, immunoreactive protein and CYP mRNA levels from human hepatocytes in primary culture versus cells treated with vehicle (0.5% methanol) and with rifampicin (rifampin) as a positive control. Rifampin 288-296 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). Nitrogen 40-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 14631602-5 2003 Interindividual variability of plasma concentration of TCA"s is connected with age, concomitant diseases, genetically determined polimorphism of cytochrome P-450 enzymes (e.g. CYP2D6) and co-medications (the drug may change pharmacokinetic properties of TCA"s). Trichloroacetic Acid 55-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-161 14631602-5 2003 Interindividual variability of plasma concentration of TCA"s is connected with age, concomitant diseases, genetically determined polimorphism of cytochrome P-450 enzymes (e.g. CYP2D6) and co-medications (the drug may change pharmacokinetic properties of TCA"s). Trichloroacetic Acid 254-257 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-161 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). Nitrogen 40-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). desethylchloroquine 67-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). desethylchloroquine 67-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). desethylchloroquine 88-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). desethylchloroquine 88-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). N,N-dideethylchloroquine 97-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). N,N-dideethylchloroquine 97-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). N,N-dideethylchloroquine 121-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-146 12967198-2 2003 It is metabolized in humans through the N-dealkylation pathway, to desethylchloroquine (DCQ) and bisdesethylchloroquine (BDCQ), by cytochrome P450 (CYP). N,N-dideethylchloroquine 121-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 12865317-0 2003 Characterization of the oxidative metabolites of 17beta-estradiol and estrone formed by 15 selectively expressed human cytochrome p450 isoforms. Estradiol 49-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-134 12908908-4 2003 DMBA is metabolized into a DNA-attacking moiety by two phase I cytochrome P450 (CYP) enzymes CYP1A1 and CYP1B1. 6,11-dimethylbenzo(b)naphtho(2,3-d)thiophene 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 12908908-4 2003 DMBA is metabolized into a DNA-attacking moiety by two phase I cytochrome P450 (CYP) enzymes CYP1A1 and CYP1B1. 6,11-dimethylbenzo(b)naphtho(2,3-d)thiophene 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 12865317-14 2003 In conclusion, many human CYP isoforms are involved in the oxidative metabolism of 17beta-estradiol and estrone, with a varying degree of catalytic activity and distinct regioselectivity. Estradiol 83-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 12865317-0 2003 Characterization of the oxidative metabolites of 17beta-estradiol and estrone formed by 15 selectively expressed human cytochrome p450 isoforms. Estrone 70-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-134 12865317-14 2003 In conclusion, many human CYP isoforms are involved in the oxidative metabolism of 17beta-estradiol and estrone, with a varying degree of catalytic activity and distinct regioselectivity. Estrone 104-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 12865317-1 2003 We systematically characterized the oxidative metabolites of 17beta-estradiol and estrone formed by 15 human cytochrome P450 (CYP) isoforms. Estradiol 61-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 12865317-1 2003 We systematically characterized the oxidative metabolites of 17beta-estradiol and estrone formed by 15 human cytochrome P450 (CYP) isoforms. Estradiol 61-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 12865317-1 2003 We systematically characterized the oxidative metabolites of 17beta-estradiol and estrone formed by 15 human cytochrome P450 (CYP) isoforms. Estrone 82-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 12865317-1 2003 We systematically characterized the oxidative metabolites of 17beta-estradiol and estrone formed by 15 human cytochrome P450 (CYP) isoforms. Estrone 82-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 12936704-0 2003 Identification of CYP3A4 and CYP2C8 as the major cytochrome P450 s responsible for morphine N-demethylation in human liver microsomes. Morphine 83-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 12936704-0 2003 Identification of CYP3A4 and CYP2C8 as the major cytochrome P450 s responsible for morphine N-demethylation in human liver microsomes. Nitrogen 92-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 12936704-2 2003 The aim was to identify the cytochrome P450 (CYP) enzymes responsible for the N-demethylation of morphine in vitro. Nitrogen 78-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 12936704-2 2003 The aim was to identify the cytochrome P450 (CYP) enzymes responsible for the N-demethylation of morphine in vitro. Nitrogen 78-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 12936704-2 2003 The aim was to identify the cytochrome P450 (CYP) enzymes responsible for the N-demethylation of morphine in vitro. Morphine 97-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 12936704-2 2003 The aim was to identify the cytochrome P450 (CYP) enzymes responsible for the N-demethylation of morphine in vitro. Morphine 97-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 12936704-5 2003 In microsomes from a panel of 14 human livers phenotyped for 10 CYP enzymes, morphine N-demethylation correlated with testosterone 6beta-hydroxylation (r=0.91, p<0.001) and paclitaxel 6-alpha hydroxylation (r=0.72, p<0.001), two specific markers of CYP3A4 and CYP2C8, respectively. morphine n 77-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 12936704-5 2003 In microsomes from a panel of 14 human livers phenotyped for 10 CYP enzymes, morphine N-demethylation correlated with testosterone 6beta-hydroxylation (r=0.91, p<0.001) and paclitaxel 6-alpha hydroxylation (r=0.72, p<0.001), two specific markers of CYP3A4 and CYP2C8, respectively. Testosterone 118-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 12893130-1 2003 Tramadol is an opioid drug metabolised in phase I by cytochrome P450 (CYP) enzymes, of which CYP2D6 is mainly responsible for the O-demethylation of tramadol, but is not involved in N-demethylation. Tramadol 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 12893130-1 2003 Tramadol is an opioid drug metabolised in phase I by cytochrome P450 (CYP) enzymes, of which CYP2D6 is mainly responsible for the O-demethylation of tramadol, but is not involved in N-demethylation. Nitrogen 182-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 14582238-4 2003 Genetic polymorphisms that influence human genotoxic risk are those of glutathione s-transferase and cytochrome P450 in exposure to polycyclic aromatic hydrocarbons (PAHs), those of N-acetyltransferase in both occupational and environmental exposures to aromatic amines (AAs) and similar compounds. Polycyclic Aromatic Hydrocarbons 132-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 12818727-6 2003 epsilon-viniferin displayed a more potent inhibitory effect than resveratrol for all the CYP activities tested (Ki 0.5 to 20 microM vs. 10 to 100 microM, respectively). epsilon-viniferin 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 12818727-6 2003 epsilon-viniferin displayed a more potent inhibitory effect than resveratrol for all the CYP activities tested (Ki 0.5 to 20 microM vs. 10 to 100 microM, respectively). Resveratrol 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 12818727-10 2003 It displayed, like resveratrol, mixed-type inhibitions for all the CYP tested, except for CYP2E1 (non-competitive). Resveratrol 19-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 12818727-11 2003 Comparison of the inhibitory effects exerted on CYP activities by epsilon-viniferin, resveratrol and non volatile components from red wine or various Cognac beverages showed that neither resveratrol, nor epsilon-viniferin is the main CYP inhibitor present in red wine solids. epsilon-viniferin 66-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 12818727-11 2003 Comparison of the inhibitory effects exerted on CYP activities by epsilon-viniferin, resveratrol and non volatile components from red wine or various Cognac beverages showed that neither resveratrol, nor epsilon-viniferin is the main CYP inhibitor present in red wine solids. Resveratrol 85-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 12893130-1 2003 Tramadol is an opioid drug metabolised in phase I by cytochrome P450 (CYP) enzymes, of which CYP2D6 is mainly responsible for the O-demethylation of tramadol, but is not involved in N-demethylation. Tramadol 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 12893130-1 2003 Tramadol is an opioid drug metabolised in phase I by cytochrome P450 (CYP) enzymes, of which CYP2D6 is mainly responsible for the O-demethylation of tramadol, but is not involved in N-demethylation. Tramadol 149-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 12893130-1 2003 Tramadol is an opioid drug metabolised in phase I by cytochrome P450 (CYP) enzymes, of which CYP2D6 is mainly responsible for the O-demethylation of tramadol, but is not involved in N-demethylation. Tramadol 149-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 12818727-0 2003 Differential inhibition of human cytochrome P450 enzymes by epsilon-viniferin, the dimer of resveratrol: comparison with resveratrol and polyphenols from alcoholized beverages. epsilon-viniferin 60-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 12818727-0 2003 Differential inhibition of human cytochrome P450 enzymes by epsilon-viniferin, the dimer of resveratrol: comparison with resveratrol and polyphenols from alcoholized beverages. Resveratrol 92-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-48 12818727-2 2003 As resveratrol and polyphenols from red wine were reported to inhibit cytochrome P450 (CYP) activities, this led us to investigate the inhibitory effects of epsilon-viniferin on human CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2B6, CYP2E1, CYP3A4 and CYP4A activities. Resveratrol 3-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 12818727-2 2003 As resveratrol and polyphenols from red wine were reported to inhibit cytochrome P450 (CYP) activities, this led us to investigate the inhibitory effects of epsilon-viniferin on human CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2B6, CYP2E1, CYP3A4 and CYP4A activities. Resveratrol 3-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 12818727-2 2003 As resveratrol and polyphenols from red wine were reported to inhibit cytochrome P450 (CYP) activities, this led us to investigate the inhibitory effects of epsilon-viniferin on human CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2B6, CYP2E1, CYP3A4 and CYP4A activities. Polyphenols 19-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 12818727-2 2003 As resveratrol and polyphenols from red wine were reported to inhibit cytochrome P450 (CYP) activities, this led us to investigate the inhibitory effects of epsilon-viniferin on human CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2B6, CYP2E1, CYP3A4 and CYP4A activities. Polyphenols 19-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 12848784-1 2003 AIMS: To identify the cytochrome P450 (CYP) isoforms responsible for the metabolism of simvastatin hydroxy acid (SVA), the most potent metabolite of simvastatin (SV). simvastatin hydroxyacid 87-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 12848784-1 2003 AIMS: To identify the cytochrome P450 (CYP) isoforms responsible for the metabolism of simvastatin hydroxy acid (SVA), the most potent metabolite of simvastatin (SV). simvastatin hydroxyacid 87-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 12848784-1 2003 AIMS: To identify the cytochrome P450 (CYP) isoforms responsible for the metabolism of simvastatin hydroxy acid (SVA), the most potent metabolite of simvastatin (SV). simvastatin hydroxyacid 113-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 12848784-1 2003 AIMS: To identify the cytochrome P450 (CYP) isoforms responsible for the metabolism of simvastatin hydroxy acid (SVA), the most potent metabolite of simvastatin (SV). simvastatin hydroxyacid 113-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 12848784-1 2003 AIMS: To identify the cytochrome P450 (CYP) isoforms responsible for the metabolism of simvastatin hydroxy acid (SVA), the most potent metabolite of simvastatin (SV). Simvastatin 87-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 12848784-1 2003 AIMS: To identify the cytochrome P450 (CYP) isoforms responsible for the metabolism of simvastatin hydroxy acid (SVA), the most potent metabolite of simvastatin (SV). Simvastatin 87-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 14531067-0 2003 An adamantanone derivative that is an original modulator of redox processes in the cytochrome P-450 system can serve as an effective remedy against obliterating angiopathy of lower extremities. adamantanone 3-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 14582238-4 2003 Genetic polymorphisms that influence human genotoxic risk are those of glutathione s-transferase and cytochrome P450 in exposure to polycyclic aromatic hydrocarbons (PAHs), those of N-acetyltransferase in both occupational and environmental exposures to aromatic amines (AAs) and similar compounds. Polycyclic Aromatic Hydrocarbons 166-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 14582238-4 2003 Genetic polymorphisms that influence human genotoxic risk are those of glutathione s-transferase and cytochrome P450 in exposure to polycyclic aromatic hydrocarbons (PAHs), those of N-acetyltransferase in both occupational and environmental exposures to aromatic amines (AAs) and similar compounds. aromatic amines 254-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 14582238-4 2003 Genetic polymorphisms that influence human genotoxic risk are those of glutathione s-transferase and cytochrome P450 in exposure to polycyclic aromatic hydrocarbons (PAHs), those of N-acetyltransferase in both occupational and environmental exposures to aromatic amines (AAs) and similar compounds. aas 271-274 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 14581727-0 2003 Effects of cytochrome P-450 inducers on the perazine metabolism in a primary culture of human hepatocytes. Perazine 44-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-27 12728288-1 2003 BACKGROUND: In patients on oral anticoagulation with warfarin, genetic variations of the cytochrome P 450-CYP2C9 have recently been associated with very low warfarin requirements. Warfarin 53-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-112 12728288-1 2003 BACKGROUND: In patients on oral anticoagulation with warfarin, genetic variations of the cytochrome P 450-CYP2C9 have recently been associated with very low warfarin requirements. Warfarin 157-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-112 12849118-2 2003 Many drugs are metabolised by the hepatic cytochrome P450 (CYP) isoenzyme system, and coadministration of AEDs and CTDs can lead to clinically relevant interactions by induction or inhibition of enzymes in shared metabolic pathways. beta-cyclodextrin tetradecasulfate 115-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 12849118-2 2003 Many drugs are metabolised by the hepatic cytochrome P450 (CYP) isoenzyme system, and coadministration of AEDs and CTDs can lead to clinically relevant interactions by induction or inhibition of enzymes in shared metabolic pathways. beta-cyclodextrin tetradecasulfate 115-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 12763054-1 2003 Tocopherols and tocotrienols are metabolized by side chain degradation initiated by cytochrome P450 (CYP)-catalyzed omega-hydroxylation followed by beta-oxidation. Tocopherols 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 12763054-1 2003 Tocopherols and tocotrienols are metabolized by side chain degradation initiated by cytochrome P450 (CYP)-catalyzed omega-hydroxylation followed by beta-oxidation. Tocopherols 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 12763054-1 2003 Tocopherols and tocotrienols are metabolized by side chain degradation initiated by cytochrome P450 (CYP)-catalyzed omega-hydroxylation followed by beta-oxidation. Tocotrienols 16-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-99 12763054-1 2003 Tocopherols and tocotrienols are metabolized by side chain degradation initiated by cytochrome P450 (CYP)-catalyzed omega-hydroxylation followed by beta-oxidation. Tocotrienols 16-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 12698250-1 2003 OBJECTIVE: Antipyrine metabolism is a "gold standard" measure of mixed cytochrome P450 (CYP) mediated drug metabolism in humans. Antipyrine 11-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 12814453-1 2003 AIMS: Nafcillin (Wyeth Laboratories, Philadelphia, PA, USA) has been reported to induce the metabolism of cyclosporin and warfarin, which are known substrates of cytochrome P-450 (CYP). Warfarin 122-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 12814453-7 2003 CONCLUSIONS: The results show that nafcillin pretreatment markedly increased the clearance of nifedipine and suggest that nafcillin is a potent inducer of CYP enzyme. Nafcillin 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-158 12814453-7 2003 CONCLUSIONS: The results show that nafcillin pretreatment markedly increased the clearance of nifedipine and suggest that nafcillin is a potent inducer of CYP enzyme. Nafcillin 122-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-158 12814453-1 2003 AIMS: Nafcillin (Wyeth Laboratories, Philadelphia, PA, USA) has been reported to induce the metabolism of cyclosporin and warfarin, which are known substrates of cytochrome P-450 (CYP). Nafcillin 6-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-178 12814453-1 2003 AIMS: Nafcillin (Wyeth Laboratories, Philadelphia, PA, USA) has been reported to induce the metabolism of cyclosporin and warfarin, which are known substrates of cytochrome P-450 (CYP). Nafcillin 6-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 12814453-1 2003 AIMS: Nafcillin (Wyeth Laboratories, Philadelphia, PA, USA) has been reported to induce the metabolism of cyclosporin and warfarin, which are known substrates of cytochrome P-450 (CYP). Cyclosporine 106-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-178 12814453-1 2003 AIMS: Nafcillin (Wyeth Laboratories, Philadelphia, PA, USA) has been reported to induce the metabolism of cyclosporin and warfarin, which are known substrates of cytochrome P-450 (CYP). Cyclosporine 106-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 12814453-1 2003 AIMS: Nafcillin (Wyeth Laboratories, Philadelphia, PA, USA) has been reported to induce the metabolism of cyclosporin and warfarin, which are known substrates of cytochrome P-450 (CYP). Warfarin 122-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-178 12698250-1 2003 OBJECTIVE: Antipyrine metabolism is a "gold standard" measure of mixed cytochrome P450 (CYP) mediated drug metabolism in humans. Antipyrine 11-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 12698250-2 2003 Cytokines (e.g., interleukin-6) and nitric oxide reduce CYP 450 activity in vitro and in vivo. Nitric Oxide 36-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 12698250-3 2003 Because interleukin-6 and nitric oxide production increases in children with sepsis-induced multiple organ failure, we hypothesized impaired CYP 450 mediated drug metabolism in this population. Nitric Oxide 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-144 12870664-0 2003 Effects of cytochrome P450 inhibitors on agonist-induced Ca2+ responses and production of NO and PGI2 in vascular endothelial cells. Epoprostenol 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-26 12930004-3 2003 PAHs have been well established as an enzyme inducer of cytochrome P450 (CYP) such as CYP1A1 and CYP1A2. Polycyclic Aromatic Hydrocarbons 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 12930004-3 2003 PAHs have been well established as an enzyme inducer of cytochrome P450 (CYP) such as CYP1A1 and CYP1A2. Polycyclic Aromatic Hydrocarbons 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 12870655-0 2003 Transcript profiling of cytochrome P450 genes in HL-60 human leukemic cells: upregulation of CYP1B1 by all-trans-retinoic acid. Tretinoin 103-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 12870664-2 2003 Cytochrome P450 (CYP), shown to mediate endothelium-dependent hyperpolarization via epoxyeicosatrienoic acids, is one of the candidates for EDHF. epoxyeicosatrienoic acids 84-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12870655-2 2003 The biotransformation of this drug is catalyzed by various cytochrome P450 (CYP) enzymes, but relatively little is known about the effect of ATRA on CYP enzyme expression in leukemic cells. Tretinoin 141-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-152 12870664-2 2003 Cytochrome P450 (CYP), shown to mediate endothelium-dependent hyperpolarization via epoxyeicosatrienoic acids, is one of the candidates for EDHF. epoxyeicosatrienoic acids 84-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 12870664-2 2003 Cytochrome P450 (CYP), shown to mediate endothelium-dependent hyperpolarization via epoxyeicosatrienoic acids, is one of the candidates for EDHF. edhf 140-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12870664-2 2003 Cytochrome P450 (CYP), shown to mediate endothelium-dependent hyperpolarization via epoxyeicosatrienoic acids, is one of the candidates for EDHF. edhf 140-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 12870664-10 2003 These data suggest that, in addition to its regulation of EDHF production, CYP also contributes to the regulation of other endothelium-dependent vasorelaxing factors by modifying EC Ca2+ signaling. edhf 58-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 12819521-4 2003 Cytochrome P450 activity was assessed by measuring the urine 6 beta-hydroxycortisol to cortisol ratio (6 beta-OHF/F). beta-hydroxycortisol 63-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12819521-4 2003 Cytochrome P450 activity was assessed by measuring the urine 6 beta-hydroxycortisol to cortisol ratio (6 beta-OHF/F). Hydrocortisone 75-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12819521-4 2003 Cytochrome P450 activity was assessed by measuring the urine 6 beta-hydroxycortisol to cortisol ratio (6 beta-OHF/F). beta-ohf 105-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12819521-4 2003 Cytochrome P450 activity was assessed by measuring the urine 6 beta-hydroxycortisol to cortisol ratio (6 beta-OHF/F). Fluorine 112-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12785830-1 2003 Intramolecular kinetic isotope effects (KIEs) were determined for cytochrome P450-catalyzed hydroxylation reactions of methyl-dideuterated trans-2-phenylcyclopropylmethane-d2 (1-d2), which gives two products from oxidation of the methyl group, trans-2-phenylcyclopropylmethanol (2) and 1-phenyl-3-buten-1ol (3). trans-2-phenylcyclopropylmethane-d2 139-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 12785830-1 2003 Intramolecular kinetic isotope effects (KIEs) were determined for cytochrome P450-catalyzed hydroxylation reactions of methyl-dideuterated trans-2-phenylcyclopropylmethane-d2 (1-d2), which gives two products from oxidation of the methyl group, trans-2-phenylcyclopropylmethanol (2) and 1-phenyl-3-buten-1ol (3). 1-d2 176-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 12785830-1 2003 Intramolecular kinetic isotope effects (KIEs) were determined for cytochrome P450-catalyzed hydroxylation reactions of methyl-dideuterated trans-2-phenylcyclopropylmethane-d2 (1-d2), which gives two products from oxidation of the methyl group, trans-2-phenylcyclopropylmethanol (2) and 1-phenyl-3-buten-1ol (3). trans-2-phenylcyclopropylmethanol 244-277 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 12785830-1 2003 Intramolecular kinetic isotope effects (KIEs) were determined for cytochrome P450-catalyzed hydroxylation reactions of methyl-dideuterated trans-2-phenylcyclopropylmethane-d2 (1-d2), which gives two products from oxidation of the methyl group, trans-2-phenylcyclopropylmethanol (2) and 1-phenyl-3-buten-1ol (3). 1-phenyl-3-buten-1ol 286-306 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 12867215-12 2003 However, as 80% of voriconazole is hepatically eliminated, primarily via the cytochrome P450 (CYP) isozymes CYP2C19, CYP3A4, and CYP2C9, voriconazole has a high potential for drug interactions, and dose reduction is recommended in patients with mild to moderate hepatic dysfunction (Child-Pugh class A or B). Voriconazole 19-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 12787491-0 2003 Microplate assay measurement of cytochrome p450-carbon monoxide complexes. Carbon Monoxide 48-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 12867215-12 2003 However, as 80% of voriconazole is hepatically eliminated, primarily via the cytochrome P450 (CYP) isozymes CYP2C19, CYP3A4, and CYP2C9, voriconazole has a high potential for drug interactions, and dose reduction is recommended in patients with mild to moderate hepatic dysfunction (Child-Pugh class A or B). Voriconazole 19-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 12867215-12 2003 However, as 80% of voriconazole is hepatically eliminated, primarily via the cytochrome P450 (CYP) isozymes CYP2C19, CYP3A4, and CYP2C9, voriconazole has a high potential for drug interactions, and dose reduction is recommended in patients with mild to moderate hepatic dysfunction (Child-Pugh class A or B). Voriconazole 137-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 12867215-12 2003 However, as 80% of voriconazole is hepatically eliminated, primarily via the cytochrome P450 (CYP) isozymes CYP2C19, CYP3A4, and CYP2C9, voriconazole has a high potential for drug interactions, and dose reduction is recommended in patients with mild to moderate hepatic dysfunction (Child-Pugh class A or B). Voriconazole 137-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 12695341-0 2003 Identification of the cytochrome P450 enzymes involved in the N-oxidation of voriconazole. Voriconazole 77-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 12752938-0 2003 Cytotoxicity of bisphenol A glycidyl methacrylate on cytochrome P450-producing cells. Bisphenol A-Glycidyl Methacrylate 16-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 12959412-1 2003 Many administered drugs are first activated by phase I drug-metabolizing enzymes, such as cytochrome P450 (CYP), and then conjugated with ligands such as UDPGA, PAPS, and glutathione by phase II drug-metabolizing enzymes, and finally excreted by transporters. Phosphoadenosine Phosphosulfate 161-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 12959412-1 2003 Many administered drugs are first activated by phase I drug-metabolizing enzymes, such as cytochrome P450 (CYP), and then conjugated with ligands such as UDPGA, PAPS, and glutathione by phase II drug-metabolizing enzymes, and finally excreted by transporters. Phosphoadenosine Phosphosulfate 161-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-110 12959412-1 2003 Many administered drugs are first activated by phase I drug-metabolizing enzymes, such as cytochrome P450 (CYP), and then conjugated with ligands such as UDPGA, PAPS, and glutathione by phase II drug-metabolizing enzymes, and finally excreted by transporters. Glutathione 171-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 12959412-1 2003 Many administered drugs are first activated by phase I drug-metabolizing enzymes, such as cytochrome P450 (CYP), and then conjugated with ligands such as UDPGA, PAPS, and glutathione by phase II drug-metabolizing enzymes, and finally excreted by transporters. Glutathione 171-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-110 12759628-3 2003 The effects of ATRA can be potentiated by inhibition of cytochrome P-450, which is known to occur with certain drugs. Tretinoin 15-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 12570834-7 2003 Another important way of demethylation generated formaldehyde production is given by microsomal cytochrome P-450 dependent oxidation of xenobiotics, such as various drugs prescribed by doctors. Formaldehyde 49-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 26984794-2 2003 OBJECTIVE: This study investigates whether low serum folate levels may contribute to depressive mood and difficulties in mental processing in patients with epilepsy treated with anti-epileptic drugs inducing the cytochrome P450. Folic Acid 53-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 212-227 12725870-1 2003 Microsomal P450-mediated monooxygenase activity supported by NADPH requires an interaction between flavoprotein NADPH-cytochrome P450 reductase and cytochrome P450. NADP 61-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 12586744-1 2003 Cytochrome P450 (CYP) epoxygenase products, such as 11,12-epoxyeicosatrienoic acid (EET), stimulate endothelial cell proliferation. 11,12-epoxy-5,8,14-eicosatrienoic acid 52-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12721102-0 2003 Contribution of human cytochrome p-450 isoforms to the metabolism of the simplest phenothiazine neuroleptic promazine. phenothiazine 82-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 12721102-0 2003 Contribution of human cytochrome p-450 isoforms to the metabolism of the simplest phenothiazine neuroleptic promazine. Promazine 108-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 12721102-2 2003 The aim of the present study was to identify human cytochrome p-450 isoforms (CYPs) involved in 5-sulphoxidation and N-demethylation of the simplest phenothiazine neuroleptic promazine in human liver. phenothiazine 149-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 12721102-2 2003 The aim of the present study was to identify human cytochrome p-450 isoforms (CYPs) involved in 5-sulphoxidation and N-demethylation of the simplest phenothiazine neuroleptic promazine in human liver. Promazine 175-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 12586744-1 2003 Cytochrome P450 (CYP) epoxygenase products, such as 11,12-epoxyeicosatrienoic acid (EET), stimulate endothelial cell proliferation. 11,12-epoxy-5,8,14-eicosatrienoic acid 52-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 12586744-1 2003 Cytochrome P450 (CYP) epoxygenase products, such as 11,12-epoxyeicosatrienoic acid (EET), stimulate endothelial cell proliferation. 11,12-epoxy-5,8,14-eicosatrienoic acid 84-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12586744-1 2003 Cytochrome P450 (CYP) epoxygenase products, such as 11,12-epoxyeicosatrienoic acid (EET), stimulate endothelial cell proliferation. 11,12-epoxy-5,8,14-eicosatrienoic acid 84-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 12599212-0 2003 Cyclooxygenase and cytochrome P-450 pathways induced by fetal calf serum regulate wound closure in 3T6 fibroblast cultures through the effect of prostaglandin E2 and 12 and 20 hydroxyeicosatetraenoic acids. Dinoprostone 145-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 12599212-0 2003 Cyclooxygenase and cytochrome P-450 pathways induced by fetal calf serum regulate wound closure in 3T6 fibroblast cultures through the effect of prostaglandin E2 and 12 and 20 hydroxyeicosatetraenoic acids. Hydroxyeicosatetraenoic Acids 176-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 12599212-6 2003 On the other hand, we show that ketoconazole, a cytochrome P-450 inhibitor, hinders the wound closure induced by FCS in wounded 3T6 fibroblast cultures. Ketoconazole 32-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 12599212-7 2003 12 and 20 Hydroxyeicosatetraenoic acids (HETEs), which are key AA metabolites synthesized by cytochrome P-450, partially revert the effects of ketoconazole on the wound repair process. Hydroxyeicosatetraenoic Acids 10-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 12599212-7 2003 12 and 20 Hydroxyeicosatetraenoic acids (HETEs), which are key AA metabolites synthesized by cytochrome P-450, partially revert the effects of ketoconazole on the wound repair process. Ketoconazole 143-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 12599212-8 2003 Thus, the COX and cytochrome P-450 pathways of the arachidonate cascade are involved in 3T6 fibroblast wound closure. Arachidonic Acid 51-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 12739762-4 2003 Then we tested the effects of phenobarbital and cyclophosphamide on CYP expression in both models. Phenobarbital 30-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 12698306-1 2003 OBJECTIVE: To screen for inhibitory effects of diosmin on cytochrome P(450)-mediated metabolism of metronidazole in healthy volunteers. Diosmin 47-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-75 12698306-1 2003 OBJECTIVE: To screen for inhibitory effects of diosmin on cytochrome P(450)-mediated metabolism of metronidazole in healthy volunteers. Metronidazole 99-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-75 12739762-4 2003 Then we tested the effects of phenobarbital and cyclophosphamide on CYP expression in both models. Cyclophosphamide 48-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 19475278-6 2003 Cytochrome P450 isoenzymes show genetic polymorphisms in neuropsychotropic drugs metabolism and the slow acetylation of N-acetyltransferase in equatorial populations (95%) increases isoniazid and hydrazine toxicity. Isoniazid 182-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 19475278-6 2003 Cytochrome P450 isoenzymes show genetic polymorphisms in neuropsychotropic drugs metabolism and the slow acetylation of N-acetyltransferase in equatorial populations (95%) increases isoniazid and hydrazine toxicity. hydrazine 196-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12388057-6 2003 We found that in high endothelial venular cells (SVEC4-10), multiple inhibitors of CYP450 monooxygenases (SKF-525a, ketoconazole, troleandomycin, itraconazole) attenuated TNF-alpha induction of MAdCAM-1, whereas NADPH oxidase inhibition (PR-39) did not. svec4-10 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-89 12734935-0 2003 [Stereoselective determination of propranolol enantiomer in transgenic cell lines expressing human cytochrome P450]. Propranolol 34-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-114 12734935-1 2003 OBJECTIVE: To establish a chiro chromatography for studying the stereoselective metabolism of propranolol (PL) in S(9) incubates prepared from transgenic cell lines expressing human cytochrome P450. Propranolol 94-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-197 12734935-1 2003 OBJECTIVE: To establish a chiro chromatography for studying the stereoselective metabolism of propranolol (PL) in S(9) incubates prepared from transgenic cell lines expressing human cytochrome P450. Propranolol 107-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-197 12617777-1 2003 AIM: To study the metabolism of cilnidipine and the effects of selective cytochrome P-450 (CYP450) inhibitors on the metabolism of cilnidipine in human liver microsomes in vitro. cilnidipine 131-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 12617777-1 2003 AIM: To study the metabolism of cilnidipine and the effects of selective cytochrome P-450 (CYP450) inhibitors on the metabolism of cilnidipine in human liver microsomes in vitro. cilnidipine 131-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-97 12617777-3 2003 Various selective CYP450 inhibitors were used to investigate their effects on the metabolism of cilnidipine and the principal CYP450 isoform involved in dehydrogenation of dihydropyridine ring of cilnidipine in human liver microsomes. cilnidipine 96-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-24 12617777-3 2003 Various selective CYP450 inhibitors were used to investigate their effects on the metabolism of cilnidipine and the principal CYP450 isoform involved in dehydrogenation of dihydropyridine ring of cilnidipine in human liver microsomes. 1,4-dihydropyridine 172-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-24 12617777-3 2003 Various selective CYP450 inhibitors were used to investigate their effects on the metabolism of cilnidipine and the principal CYP450 isoform involved in dehydrogenation of dihydropyridine ring of cilnidipine in human liver microsomes. 1,4-dihydropyridine 172-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-132 12617777-3 2003 Various selective CYP450 inhibitors were used to investigate their effects on the metabolism of cilnidipine and the principal CYP450 isoform involved in dehydrogenation of dihydropyridine ring of cilnidipine in human liver microsomes. cilnidipine 196-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-24 12617777-3 2003 Various selective CYP450 inhibitors were used to investigate their effects on the metabolism of cilnidipine and the principal CYP450 isoform involved in dehydrogenation of dihydropyridine ring of cilnidipine in human liver microsomes. cilnidipine 196-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-132 12564927-1 2003 Human cytochrome P450 (P450) 2D6 is an important enzyme involved in the metabolism of drugs, many of which are amines or contain other basic nitrogen atoms. Amines 111-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-32 12564927-1 2003 Human cytochrome P450 (P450) 2D6 is an important enzyme involved in the metabolism of drugs, many of which are amines or contain other basic nitrogen atoms. Nitrogen 141-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-32 12637244-0 2003 Characterization of the cytochrome P450 enzymes involved in the in vitro metabolism of ethosuximide by human hepatic microsomal enzymes. Ethosuximide 87-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 12637244-2 2003 The relative roles of human hepatic cytochrome P450 (CYP) subfamilies participating in ethosuximide metabolism have been studied in vitro using humanized heterologous CYP microsomal systems expressing either CYP1A2, CYP2A6, CYP2C9, CYP2D6, CYP2E1 or CYP3A4. Ethosuximide 87-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 12637244-2 2003 The relative roles of human hepatic cytochrome P450 (CYP) subfamilies participating in ethosuximide metabolism have been studied in vitro using humanized heterologous CYP microsomal systems expressing either CYP1A2, CYP2A6, CYP2C9, CYP2D6, CYP2E1 or CYP3A4. Ethosuximide 87-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 12573493-3 2003 Moreover, CYP epoxygenase products as well as CYP-derived reactive oxygen species are intracellular signal transduction molecules involved in several signaling cascades affecting numerous cellular processes, including vascular cell proliferation and angiogenesis. Reactive Oxygen Species 58-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 12388057-6 2003 We found that in high endothelial venular cells (SVEC4-10), multiple inhibitors of CYP450 monooxygenases (SKF-525a, ketoconazole, troleandomycin, itraconazole) attenuated TNF-alpha induction of MAdCAM-1, whereas NADPH oxidase inhibition (PR-39) did not. Proadifen 106-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-89 12388057-6 2003 We found that in high endothelial venular cells (SVEC4-10), multiple inhibitors of CYP450 monooxygenases (SKF-525a, ketoconazole, troleandomycin, itraconazole) attenuated TNF-alpha induction of MAdCAM-1, whereas NADPH oxidase inhibition (PR-39) did not. Ketoconazole 116-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-89 12388057-6 2003 We found that in high endothelial venular cells (SVEC4-10), multiple inhibitors of CYP450 monooxygenases (SKF-525a, ketoconazole, troleandomycin, itraconazole) attenuated TNF-alpha induction of MAdCAM-1, whereas NADPH oxidase inhibition (PR-39) did not. Itraconazole 146-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-89 12653348-5 2003 Cytochrome P450 (CYP) activity was studied before and after cold storage by means of monoethylglycinexylide (MEGX) detection in the effluent medium, after repeated lidocaine injections. monoethylglycinexylide 85-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 12505783-6 2003 In the present article, we investigated by HPLC and GC-MS whether cis-EODA is formed enzymatically from oleic acid by the cytochrome P450 (CYP) system. 9,10-epoxystearic acid 66-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 12505783-6 2003 In the present article, we investigated by HPLC and GC-MS whether cis-EODA is formed enzymatically from oleic acid by the cytochrome P450 (CYP) system. 9,10-epoxystearic acid 66-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 12505783-6 2003 In the present article, we investigated by HPLC and GC-MS whether cis-EODA is formed enzymatically from oleic acid by the cytochrome P450 (CYP) system. Oleic Acid 104-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 12505783-6 2003 In the present article, we investigated by HPLC and GC-MS whether cis-EODA is formed enzymatically from oleic acid by the cytochrome P450 (CYP) system. Oleic Acid 104-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 12505783-11 2003 The epoxidation of oleic acid was found to depend on CYP, NADPH+H(+), and O(2). Oleic Acid 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 12505783-16 2003 Our study suggests that circulating and excretory cis-EODA may originate, at least in part, from CYP-catalyzed epoxidation of oleic acid. Oleic Acid 126-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids 75-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids 75-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). 14,15-epoxy-5,8,11-eicosatrienoic acid 132-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). 14,15-epoxy-5,8,11-eicosatrienoic acid 132-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). dihydroxyeicosa-trienoic acids 159-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). dihydroxyeicosa-trienoic acids 159-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). dhets 191-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). dhets 191-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 203-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 203-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 236-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12570747-1 2003 Arachidonic acid can be metabolized by cytochrome p450 (CYP450) enzymes to 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs), their corresponding dihydroxyeicosa-trienoic acids (DHETs), and 20-hydroxyeicosatetraenoic acid (20-HETE). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 236-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12570747-8 2003 CYP450 metabolites also act as second messengers for many paracrine and hormonal agents, including endothelin, nitric oxide, and angiotensin II. Nitric Oxide 111-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-6 12570747-9 2003 The production of kidney CYP450 arachidonic acid metabolites is altered in diabetes, pregnancy, hepatorenal syndrome, and in various models of hypertension, and it is likely that changes in this system contribute to the abnormalities in renal function that are associated with many of these conditions. Arachidonic Acid 32-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-31 12544321-0 2003 Rhabdomyolysis from cytochrome p-450 interaction of ketoconazole and simvastatin in prostate cancer. Simvastatin 69-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 12523956-1 2003 The aim of this work was to identify the form(s) of human liver cytochrome P450 (CYP) involved in the hepatic transformation of myristicin to its major metabolite, 5-allyl-1-methoxy-2,3-dihydroxybenzene. myristicin 128-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 12523956-1 2003 The aim of this work was to identify the form(s) of human liver cytochrome P450 (CYP) involved in the hepatic transformation of myristicin to its major metabolite, 5-allyl-1-methoxy-2,3-dihydroxybenzene. myristicin 128-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 12523956-1 2003 The aim of this work was to identify the form(s) of human liver cytochrome P450 (CYP) involved in the hepatic transformation of myristicin to its major metabolite, 5-allyl-1-methoxy-2,3-dihydroxybenzene. 5-allyl-1-methoxy-2,3-dihydroxybenzene 164-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 12523956-1 2003 The aim of this work was to identify the form(s) of human liver cytochrome P450 (CYP) involved in the hepatic transformation of myristicin to its major metabolite, 5-allyl-1-methoxy-2,3-dihydroxybenzene. 5-allyl-1-methoxy-2,3-dihydroxybenzene 164-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 12618402-6 2003 High-density oligonucleotide microarrays identified altered expression of 50 transcripts in response to TIGR/MYOC overexpression, including homologs of aquaporin-4 and cytochrome-P450, previously associated with glaucoma, and several proteins of unknown function. Oligonucleotides 13-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-183 12875104-6 2003 However, because PIs and all statins except pravastatin are metabolized by the cytochrome P450 (CYP) system, co-administration of these agents produces a significant risk of drug interactions and statin-induced hepatotoxicity and myopathy. Monothiopyrophosphoric acid 17-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 12548158-1 2003 Cytochrome P450 isoenzymes are known to contribute to estrone metabolism. Estrone 54-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12875104-6 2003 However, because PIs and all statins except pravastatin are metabolized by the cytochrome P450 (CYP) system, co-administration of these agents produces a significant risk of drug interactions and statin-induced hepatotoxicity and myopathy. Monothiopyrophosphoric acid 17-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 12875104-6 2003 However, because PIs and all statins except pravastatin are metabolized by the cytochrome P450 (CYP) system, co-administration of these agents produces a significant risk of drug interactions and statin-induced hepatotoxicity and myopathy. Pravastatin 44-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 12875104-6 2003 However, because PIs and all statins except pravastatin are metabolized by the cytochrome P450 (CYP) system, co-administration of these agents produces a significant risk of drug interactions and statin-induced hepatotoxicity and myopathy. Pravastatin 44-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-99 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Quetiapine Fumarate 158-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Clozapine 112-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 12537513-15 2003 Overall, the results of the interaction studies suggest that modafinil has potential to affect the pharmacokinetics of drugs that are metabolised by certain CYP enzymes. Modafinil 61-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Clozapine 112-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Olanzapine 126-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Olanzapine 126-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Clozapine 147-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Clozapine 147-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 12751920-7 2003 Clinical trials have studied potential interactions of zaleplon, zolpidem and zopiclone with the following types of drugs: cytochrome P450 (CYP) inducers (rifampicin), CYP inhibitors (azoles, ritonavir and erythromycin), histamine H(2) receptor antagonists (cimetidine and ranitidine), antidepressants, antipsychotics, antagonists of benzodiazepines and drugs causing sedation. zopiclone 78-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-138 12751920-14 2003 While triazolam and midazolam are biotransformed almost entirely via CYP3A4, the newer hypnosedatives are biotransformed by several CYP isozymes in addition to CYP3A4, resulting in CYP3A4 inhibitors and inducers having a lesser effect on their biotransformation. Triazolam 6-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 12751920-14 2003 While triazolam and midazolam are biotransformed almost entirely via CYP3A4, the newer hypnosedatives are biotransformed by several CYP isozymes in addition to CYP3A4, resulting in CYP3A4 inhibitors and inducers having a lesser effect on their biotransformation. Midazolam 20-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Quetiapine Fumarate 158-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). ziprasidone 173-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). ziprasidone 173-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Olanzapine 198-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Olanzapine 198-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 12846597-0 2003 Ximelagatran, an oral direct thrombin inhibitor, has a low potential for cytochrome P450-mediated drug-drug interactions. ximelagatran 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Risperidone 213-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-72 12846597-4 2003 OBJECTIVE: To investigate the potential of ximelagatran, the intermediates ethyl-melagatran and hydroxy-melagatran, and melagatran to inhibit the CYP system in vitro and in vivo, and the influence of three CYP substrates on the pharmacokinetics of melagatran in vivo. ximelagatran 43-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 12846597-15 2003 Together, the in vitro and in vivo studies indicate that metabolic drug-drug interactions involving the major human CYP enzymes should not be expected with ximelagatran. ximelagatran 156-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 12665390-6 2003 Atypical antipsychotics are metabolised predominantly by cytochrome p450 (CYP) isoenzymes, particularly CYP1A2 (clozapine and olanzapine), CYP3A4 (clozapine, quetiapine and ziprasidone) and CYP2D6 (olanzapine and risperidone). Risperidone 213-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 12846597-4 2003 OBJECTIVE: To investigate the potential of ximelagatran, the intermediates ethyl-melagatran and hydroxy-melagatran, and melagatran to inhibit the CYP system in vitro and in vivo, and the influence of three CYP substrates on the pharmacokinetics of melagatran in vivo. melagatran 45-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 12841918-11 2003 The drug interaction profile of voriconazole also warrants a careful evaluation of the concomitant medication, mainly due to cytochrome P450 metabolism. Voriconazole 32-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-140 12846597-5 2003 METHODS: The CYP inhibitory properties of ximelagatran, the intermediates and melagatran were tested in vitro by two different methods, using heterologously expressed enzymes or human liver microsomes. ximelagatran 42-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 12846597-5 2003 METHODS: The CYP inhibitory properties of ximelagatran, the intermediates and melagatran were tested in vitro by two different methods, using heterologously expressed enzymes or human liver microsomes. melagatran 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 14664653-9 2003 However, there are significant genetic polymorphisms for one of the cytochrome P450 (CYP) isoenzymes involved in PPI metabolism (CYP2C19), and this polymorphism has been shown to substantially increase plasma levels of omeprazole, lansoprazole and pantoprazole, but not those of rabeprazole. Omeprazole 219-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 14664653-9 2003 However, there are significant genetic polymorphisms for one of the cytochrome P450 (CYP) isoenzymes involved in PPI metabolism (CYP2C19), and this polymorphism has been shown to substantially increase plasma levels of omeprazole, lansoprazole and pantoprazole, but not those of rabeprazole. Omeprazole 219-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 14664653-9 2003 However, there are significant genetic polymorphisms for one of the cytochrome P450 (CYP) isoenzymes involved in PPI metabolism (CYP2C19), and this polymorphism has been shown to substantially increase plasma levels of omeprazole, lansoprazole and pantoprazole, but not those of rabeprazole. Lansoprazole 231-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 14664653-9 2003 However, there are significant genetic polymorphisms for one of the cytochrome P450 (CYP) isoenzymes involved in PPI metabolism (CYP2C19), and this polymorphism has been shown to substantially increase plasma levels of omeprazole, lansoprazole and pantoprazole, but not those of rabeprazole. Lansoprazole 231-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 15618733-0 2003 Metabolism of N,N-dipropyl-2-[4-methoxy-3-(2-phenyl-ethoxy)-phenyl]-ethyl-amine-monohydrochloride (NE-100), a novel sigma ligand: contribution of cytochrome P450 forms involved in the formation of individual metabolites in human liver and small intestine. N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 14-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-161 14664653-9 2003 However, there are significant genetic polymorphisms for one of the cytochrome P450 (CYP) isoenzymes involved in PPI metabolism (CYP2C19), and this polymorphism has been shown to substantially increase plasma levels of omeprazole, lansoprazole and pantoprazole, but not those of rabeprazole. Pantoprazole 248-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 15618733-0 2003 Metabolism of N,N-dipropyl-2-[4-methoxy-3-(2-phenyl-ethoxy)-phenyl]-ethyl-amine-monohydrochloride (NE-100), a novel sigma ligand: contribution of cytochrome P450 forms involved in the formation of individual metabolites in human liver and small intestine. N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 99-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-161 15618733-1 2003 In the present study, human cytochrome P450 (CYP) forms involved in producing the primary metabolites of NE-100 were identified. N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 105-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 15618733-1 2003 In the present study, human cytochrome P450 (CYP) forms involved in producing the primary metabolites of NE-100 were identified. N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 105-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 14664653-9 2003 However, there are significant genetic polymorphisms for one of the cytochrome P450 (CYP) isoenzymes involved in PPI metabolism (CYP2C19), and this polymorphism has been shown to substantially increase plasma levels of omeprazole, lansoprazole and pantoprazole, but not those of rabeprazole. Pantoprazole 248-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 15618748-0 2003 In vitro inhibitory effect of 1-aminobenzotriazole on drug oxidations catalyzed by human cytochrome P450 enzymes: a comparison with SKF-525A and ketoconazole. 1-aminobenzotriazole 30-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 15618748-1 2003 1-Aminobenzotriazole (ABT) is widely used as a non-specific inhibitor of animal cytochrome P450 (CYP). 1-aminobenzotriazole 0-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 14664653-9 2003 However, there are significant genetic polymorphisms for one of the cytochrome P450 (CYP) isoenzymes involved in PPI metabolism (CYP2C19), and this polymorphism has been shown to substantially increase plasma levels of omeprazole, lansoprazole and pantoprazole, but not those of rabeprazole. Rabeprazole 279-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 15618748-1 2003 1-Aminobenzotriazole (ABT) is widely used as a non-specific inhibitor of animal cytochrome P450 (CYP). 1-aminobenzotriazole 0-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 15618748-1 2003 1-Aminobenzotriazole (ABT) is widely used as a non-specific inhibitor of animal cytochrome P450 (CYP). 1-aminobenzotriazole 22-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 14664653-9 2003 However, there are significant genetic polymorphisms for one of the cytochrome P450 (CYP) isoenzymes involved in PPI metabolism (CYP2C19), and this polymorphism has been shown to substantially increase plasma levels of omeprazole, lansoprazole and pantoprazole, but not those of rabeprazole. Rabeprazole 279-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 15618748-1 2003 1-Aminobenzotriazole (ABT) is widely used as a non-specific inhibitor of animal cytochrome P450 (CYP). 1-aminobenzotriazole 22-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 15618748-2 2003 In the present study, the inhibitory effect of ABT was investigated on drug oxidations catalyzed by human CYP isoforms. 1-aminobenzotriazole 47-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 12569440-0 2003 Liquid chromatography/tandem mass spectrometric determination of inhibition of human cytochrome P450 isozymes by resveratrol and resveratrol-3-sulfate. Resveratrol 113-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 15618748-11 2003 ABT, SKF-525A, and ketoconazole showed different selectivity and had a wide range of Ki values for the drug oxidations catalyzed by human CYP enzymes. 1-aminobenzotriazole 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 15618748-11 2003 ABT, SKF-525A, and ketoconazole showed different selectivity and had a wide range of Ki values for the drug oxidations catalyzed by human CYP enzymes. Proadifen 5-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 15618748-11 2003 ABT, SKF-525A, and ketoconazole showed different selectivity and had a wide range of Ki values for the drug oxidations catalyzed by human CYP enzymes. Ketoconazole 19-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 15618748-12 2003 Therefore, we conclude that inhibitory studies designed to predict the contribution of CYP enzymes to the metabolism of certain compounds should be performed using multiple CYP inhibitors, such as ABT, SKF-525A, and ketoconazole. 1-aminobenzotriazole 197-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 15618748-12 2003 Therefore, we conclude that inhibitory studies designed to predict the contribution of CYP enzymes to the metabolism of certain compounds should be performed using multiple CYP inhibitors, such as ABT, SKF-525A, and ketoconazole. Proadifen 202-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 15618748-12 2003 Therefore, we conclude that inhibitory studies designed to predict the contribution of CYP enzymes to the metabolism of certain compounds should be performed using multiple CYP inhibitors, such as ABT, SKF-525A, and ketoconazole. Ketoconazole 216-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 12862505-5 2003 Losartan and irbesartan have a greater affinity for cytochrome p450 (CYP) isoenzymes and, thus, are more likely to be implicated in drug interactions. Losartan 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 12862505-5 2003 Losartan and irbesartan have a greater affinity for cytochrome p450 (CYP) isoenzymes and, thus, are more likely to be implicated in drug interactions. Losartan 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 12862505-5 2003 Losartan and irbesartan have a greater affinity for cytochrome p450 (CYP) isoenzymes and, thus, are more likely to be implicated in drug interactions. Irbesartan 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 12862505-5 2003 Losartan and irbesartan have a greater affinity for cytochrome p450 (CYP) isoenzymes and, thus, are more likely to be implicated in drug interactions. Irbesartan 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 12490585-2 2003 In the present study, we investigated the effects of arsenic trioxide (As2O3), recently used as an anticancer drug, on the expression of human cytochrome P450 (P450) 1A1, which bioactivates polycyclic aromatic hydrocarbons into mutagenic metabolites. Arsenic Trioxide 53-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-169 12490585-2 2003 In the present study, we investigated the effects of arsenic trioxide (As2O3), recently used as an anticancer drug, on the expression of human cytochrome P450 (P450) 1A1, which bioactivates polycyclic aromatic hydrocarbons into mutagenic metabolites. Arsenic Trioxide 71-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-169 12490585-2 2003 In the present study, we investigated the effects of arsenic trioxide (As2O3), recently used as an anticancer drug, on the expression of human cytochrome P450 (P450) 1A1, which bioactivates polycyclic aromatic hydrocarbons into mutagenic metabolites. Polycyclic Aromatic Hydrocarbons 190-222 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-169 12680714-0 2003 Proliferation of intracellular structure corresponding to reduced affinity of fluconazole for cytochrome P-450 in two low-susceptibility strains of Candida albicans isolated from a Japanese AIDS patient. Fluconazole 78-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 12680714-3 2003 We here examined characteristics related to cytochrome P-450 (CYP), especially sterol 14alpha-demethylase encoded by the ERG11 gene which is the target molecule for fluconazole. Fluconazole 165-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-60 12680714-3 2003 We here examined characteristics related to cytochrome P-450 (CYP), especially sterol 14alpha-demethylase encoded by the ERG11 gene which is the target molecule for fluconazole. Fluconazole 165-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 12680714-4 2003 In TIMM 3164 and 3165, the ergosterol synthesis by cell-free extracts was somewhat less susceptible to fluconazole, due to a decrease in fluconazole affinity for CYP. Ergosterol 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 12680714-4 2003 In TIMM 3164 and 3165, the ergosterol synthesis by cell-free extracts was somewhat less susceptible to fluconazole, due to a decrease in fluconazole affinity for CYP. Fluconazole 137-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 12680714-10 2003 In addition to the reduced intracellular accumulation, the decreased affinity of fluconazole for CYP in TIMM 3164 and 3165 is assumed to be associated with the fluconazole-resistance phenotype. Fluconazole 81-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 12680714-10 2003 In addition to the reduced intracellular accumulation, the decreased affinity of fluconazole for CYP in TIMM 3164 and 3165 is assumed to be associated with the fluconazole-resistance phenotype. Fluconazole 160-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 12569440-0 2003 Liquid chromatography/tandem mass spectrometric determination of inhibition of human cytochrome P450 isozymes by resveratrol and resveratrol-3-sulfate. Resveratrol 129-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 12569440-1 2003 trans-Resveratrol, a phenolic phytoalexin occurring in grapes, wine, peanuts, and cranberries, has been reported to both have anticarcinogenic, antioxidative, phytoestrogenic, and cardioprotective activities, and to be a weak inhibitor of cytochrome P450 (CYP)3A4, which might have significance for drug-drug interactions. Resveratrol 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 239-254 12569440-2 2003 Since trans-resveratrol is rapidly converted in vivo to primarily trans-resveratrol-3-sulfate, a rapid, selective, and sensitive method using liquid chromatography/tandem mass spectrometry (LC/MS/MS) was developed to investigate human cytochrome P450 inhibition by trans-resveratrol-3-sulfate. Resveratrol 6-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 235-250 12569440-3 2003 Effects of trans-resveratrol and trans-resveratrol-3-sulfate on the metabolism of selective cytochrome P450 substrates (CYP1A2/ethoxyresorufin, CYP2C9/diclofenac, CYP2C19/(S)-mephenytoin, CYP2D6/bufuralol, CYP3A4/testosterone) were monitored using cDNA-expressed human recombinant isozymes. Resveratrol 11-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 12569440-3 2003 Effects of trans-resveratrol and trans-resveratrol-3-sulfate on the metabolism of selective cytochrome P450 substrates (CYP1A2/ethoxyresorufin, CYP2C9/diclofenac, CYP2C19/(S)-mephenytoin, CYP2D6/bufuralol, CYP3A4/testosterone) were monitored using cDNA-expressed human recombinant isozymes. trans-resveratrol-3-sulfate 33-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 12426514-1 2002 BACKGROUND AND OBJECTIVE: Praziquantel is extensively metabolized by the hepatic cytochrome P450 (CYP) enzymes. Praziquantel 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 12520072-2 2003 Cytochrome P450 induction was performed by incubating lymphocytes with benzanthracene. benz(a)anthracene 71-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12469329-0 2002 Effect of zolpidem on human cytochrome P450 activity, and on transport mediated by P-glycoprotein. Zolpidem 10-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 12469329-1 2002 The influence of high concentrations of zolpidem (100 microM, corresponding to approximately 200 times maximum therapeutic concentrations) on the activity of six human Cytochrome P450 (CYP) enzymes was evaluated in a model system using human liver microsomes. Zolpidem 40-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-183 12469329-1 2002 The influence of high concentrations of zolpidem (100 microM, corresponding to approximately 200 times maximum therapeutic concentrations) on the activity of six human Cytochrome P450 (CYP) enzymes was evaluated in a model system using human liver microsomes. Zolpidem 40-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-188 12437345-0 2002 Molecular mechanism of the electron transfer reaction in cytochrome P450(cam)--putidaredoxin: roles of glutamine 360 at the heme proximal site. Glutamine 103-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-92 12437345-0 2002 Molecular mechanism of the electron transfer reaction in cytochrome P450(cam)--putidaredoxin: roles of glutamine 360 at the heme proximal site. Heme 124-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-92 12543512-1 2002 An enantioselective assay for S-(-)- and R-(+)-propranolol in transgenic Chinese hamster CHL cell lines, expressing human cytochrome P450 (CYP), was developed. s-(-)- and r-(+)-propranolol 30-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 12543512-1 2002 An enantioselective assay for S-(-)- and R-(+)-propranolol in transgenic Chinese hamster CHL cell lines, expressing human cytochrome P450 (CYP), was developed. s-(-)- and r-(+)-propranolol 30-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 12369889-11 2002 Historically, this strategy utilized CYP 2B1 to activate oxazaphosphorines. oxazaphosphorines 57-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 12426514-1 2002 BACKGROUND AND OBJECTIVE: Praziquantel is extensively metabolized by the hepatic cytochrome P450 (CYP) enzymes. Praziquantel 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. inn 10-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 15-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 12379470-3 2002 The activities of eight human recombinant cytochrome P450 (CYP) isozymes were measured in the presence of purpurin, alizarin or carminic acid. purpurin 106-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 12379470-6 2002 CYP1B1 was the most strongly affected CYP molecule by purpurin and alizarin among CYPs examined in this study. purpurin 54-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 12379470-3 2002 The activities of eight human recombinant cytochrome P450 (CYP) isozymes were measured in the presence of purpurin, alizarin or carminic acid. purpurin 106-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 12379470-6 2002 CYP1B1 was the most strongly affected CYP molecule by purpurin and alizarin among CYPs examined in this study. alizarin 67-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 12379470-3 2002 The activities of eight human recombinant cytochrome P450 (CYP) isozymes were measured in the presence of purpurin, alizarin or carminic acid. alizarin 116-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 12379470-14 2002 These results suggest that the antigenotoxic activities of purpurin and alizarin can be explained by inhibition of CYP activities responsible for activating the mutagens. purpurin 59-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 12379470-3 2002 The activities of eight human recombinant cytochrome P450 (CYP) isozymes were measured in the presence of purpurin, alizarin or carminic acid. alizarin 116-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 12379470-14 2002 These results suggest that the antigenotoxic activities of purpurin and alizarin can be explained by inhibition of CYP activities responsible for activating the mutagens. alizarin 72-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 12487729-0 2002 Comparison of rat and human cytochrome P450 (CYP) sources of N-alkylprotoporphyrin IX. n-alkylprotoporphyrin ix 61-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 12487729-0 2002 Comparison of rat and human cytochrome P450 (CYP) sources of N-alkylprotoporphyrin IX. n-alkylprotoporphyrin ix 61-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 12379470-3 2002 The activities of eight human recombinant cytochrome P450 (CYP) isozymes were measured in the presence of purpurin, alizarin or carminic acid. Carmine 128-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 12379470-3 2002 The activities of eight human recombinant cytochrome P450 (CYP) isozymes were measured in the presence of purpurin, alizarin or carminic acid. Carmine 128-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 12487729-3 2002 The porphyrinogenicity of certain xenobiotics is due to mechanism-based inactivation of selected cytochrome P450 (CYP) enzymes, with concurrent formation of N-alkylprotoporphyrins (N-alkylPPs), which disrupt control of haem biosynthesis. n-alkylprotoporphyrins 157-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 12487729-3 2002 The porphyrinogenicity of certain xenobiotics is due to mechanism-based inactivation of selected cytochrome P450 (CYP) enzymes, with concurrent formation of N-alkylprotoporphyrins (N-alkylPPs), which disrupt control of haem biosynthesis. Heme 219-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-112 12487729-3 2002 The porphyrinogenicity of certain xenobiotics is due to mechanism-based inactivation of selected cytochrome P450 (CYP) enzymes, with concurrent formation of N-alkylprotoporphyrins (N-alkylPPs), which disrupt control of haem biosynthesis. Heme 219-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 12487729-5 2002 The objective was to use cDNA-expressed individual rat CYP enzyme preparations in microsomes prepared from baculovirus-infected insect cells to determine which rat CYP enzymes were the source of N-alkylPPs after interaction with three porphyrinogenic xenobiotics and to compare the results with formation of N-alkylPPs in individual human CYP enzyme orthologues. n-alkylpps 195-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-167 12487729-7 2002 A sensitive fluorometric technique was employed to quantitate N-alkylPP formation after interaction of individual CYP enzymes with a porphyrinogenic xenobiotic. Nitrogen 62-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 12452431-1 2002 Cytochrome P450 (P450) reactions are of interest because of their relevance to the oxidative metabolism of drugs, steroids, carcinogens, and other chemicals. Steroids 114-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12487729-9 2002 N-alkylPP formation was found following the interaction of three porphyrinogenic xenobiotics with CYP1A2, 2B1, 2C6, 2C11 and 3A2, in amounts ranging from 0.45 to 0.07 nmol N-alkylPP nmol(-1) CYP. Nitrogen 0-1 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 12487729-9 2002 N-alkylPP formation was found following the interaction of three porphyrinogenic xenobiotics with CYP1A2, 2B1, 2C6, 2C11 and 3A2, in amounts ranging from 0.45 to 0.07 nmol N-alkylPP nmol(-1) CYP. alkylpp 2-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 12487729-9 2002 N-alkylPP formation was found following the interaction of three porphyrinogenic xenobiotics with CYP1A2, 2B1, 2C6, 2C11 and 3A2, in amounts ranging from 0.45 to 0.07 nmol N-alkylPP nmol(-1) CYP. n-alkylpp 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 12385721-0 2002 In vitro metabolism of carbaryl by human cytochrome P450 and its inhibition by chlorpyrifos. Carbaryl 23-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 12385721-0 2002 In vitro metabolism of carbaryl by human cytochrome P450 and its inhibition by chlorpyrifos. Chlorpyrifos 79-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 12385721-2 2002 Although previous studies have demonstrated that carbaryl can be metabolized by cytochrome P450 (CYP), the identification and characterization of CYP isoforms involved in metabolism have not been described either in humans or in experimental animals. Carbaryl 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 12385721-2 2002 Although previous studies have demonstrated that carbaryl can be metabolized by cytochrome P450 (CYP), the identification and characterization of CYP isoforms involved in metabolism have not been described either in humans or in experimental animals. Carbaryl 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 12385721-2 2002 Although previous studies have demonstrated that carbaryl can be metabolized by cytochrome P450 (CYP), the identification and characterization of CYP isoforms involved in metabolism have not been described either in humans or in experimental animals. Carbaryl 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-149 12385721-3 2002 The in vitro metabolic activities of human liver microsomes (HLM) and human cytochrome P450 (CYP) isoforms toward carbaryl were investigated in this study. Carbaryl 114-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 12385721-3 2002 The in vitro metabolic activities of human liver microsomes (HLM) and human cytochrome P450 (CYP) isoforms toward carbaryl were investigated in this study. Carbaryl 114-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 12385721-4 2002 The three major metabolites, i.e. 5-hydroxycarbaryl, 4-hydroxycarbaryl and carbaryl methylol, were identified after incubation of carbaryl with HLM or individual CYP isoforms and analysis by HPLC. (5-hydroxynaphthalen-1-yl) N-methylcarbamate 34-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 12385721-4 2002 The three major metabolites, i.e. 5-hydroxycarbaryl, 4-hydroxycarbaryl and carbaryl methylol, were identified after incubation of carbaryl with HLM or individual CYP isoforms and analysis by HPLC. Carbaryl 43-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 12385721-4 2002 The three major metabolites, i.e. 5-hydroxycarbaryl, 4-hydroxycarbaryl and carbaryl methylol, were identified after incubation of carbaryl with HLM or individual CYP isoforms and analysis by HPLC. Carbaryl 62-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 12385721-5 2002 Most of the 16 human CYP isoforms studied showed some metabolic activity toward carbaryl. Carbaryl 80-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 12385721-12 2002 Therefore, carbaryl metabolism in humans and its interaction with other chemicals is reflected by the concentration of CYP isoforms in HLM and their activities in the metabolic pathways for carbaryl. Carbaryl 11-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-122 12385721-12 2002 Therefore, carbaryl metabolism in humans and its interaction with other chemicals is reflected by the concentration of CYP isoforms in HLM and their activities in the metabolic pathways for carbaryl. Carbaryl 190-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-122 12396873-5 2002 Cotreatment of the cells with 2 microM alpha-naphthoflavone, a cytochrome P-450 inhibitor and arylhydrocarbon receptor antagonist, blocked the increase of benzo[a]pyrene hydroxylase activity induced by treatment with MEP extract alone. alpha-naphthoflavone 39-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 12351142-12 2002 Cytochrome P-450 oxidizes BZ to an inactive product (3-OHz.sbnd;BZ) and ABZ to N"HA and N-hydroxy-N-acetylbenzidine (NHA). benzidine 26-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12228186-0 2002 Inhibitory effects of tricyclic antidepressants (TCAs) on human cytochrome P450 enzymes in vitro: mechanism of drug interaction between TCAs and phenytoin. Phenytoin 145-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 12374776-1 2002 Cytochrome P450 mono-oxygenases (CYP) play an essential role in steroid metabolism, and there is speculation that sex hormones might influence cardiac mass and physiology. Steroids 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12388931-6 2002 Together these data suggest that cysteine-nitrosylation of the HNF4 DNA-binding domain is the primary molecular mechanism responsible for the drop in oxydase activities of hepatic cytochrome P450 enzymes, and the consequent impairment in drug metabolism during inflammation. Cysteine 33-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-195 12351142-12 2002 Cytochrome P-450 oxidizes BZ to an inactive product (3-OHz.sbnd;BZ) and ABZ to N"HA and N-hydroxy-N-acetylbenzidine (NHA). 3-ohz.sbnd 53-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12227954-6 2002 Skin preexposure to the skin CYP450 inducer BaP largely changed label penetration depth and distribution pattern in cutaneous tissues and decreased (14)C concentration in skin and fat. Benzo(a)pyrene 44-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-35 12351142-12 2002 Cytochrome P-450 oxidizes BZ to an inactive product (3-OHz.sbnd;BZ) and ABZ to N"HA and N-hydroxy-N-acetylbenzidine (NHA). benzidine 64-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12351142-12 2002 Cytochrome P-450 oxidizes BZ to an inactive product (3-OHz.sbnd;BZ) and ABZ to N"HA and N-hydroxy-N-acetylbenzidine (NHA). N-acetylbenzidine 72-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12351142-12 2002 Cytochrome P-450 oxidizes BZ to an inactive product (3-OHz.sbnd;BZ) and ABZ to N"HA and N-hydroxy-N-acetylbenzidine (NHA). N-hydroxy-N'-acetylbenzidine 88-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12297285-1 2002 Cytochrome P450(BSbeta) is a peroxygenase that catalyzes the alpha- or beta-hydroxylation of myristic acid by utilizing H(2)O(2). Myristic Acid 93-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-23 12297285-1 2002 Cytochrome P450(BSbeta) is a peroxygenase that catalyzes the alpha- or beta-hydroxylation of myristic acid by utilizing H(2)O(2). h(2) 120-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-23 12351142-12 2002 Cytochrome P-450 oxidizes BZ to an inactive product (3-OHz.sbnd;BZ) and ABZ to N"HA and N-hydroxy-N-acetylbenzidine (NHA). N-hydroxy-N'-acetylbenzidine 117-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12351142-13 2002 Cytochrome P-450, PHS, and horseradish peroxidase activate ABZ to bind DNA forming N"-(3"-monophospho-deoxyguanosin-8-yl)-N-acetylbenzidine (dGp-ABZ). N-acetylbenzidine 59-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12351142-13 2002 Cytochrome P-450, PHS, and horseradish peroxidase activate ABZ to bind DNA forming N"-(3"-monophospho-deoxyguanosin-8-yl)-N-acetylbenzidine (dGp-ABZ). N'-(3'-monophosphodeoxyguanosin-8-yl)-N-acetylbenzidine 83-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12351142-13 2002 Cytochrome P-450, PHS, and horseradish peroxidase activate ABZ to bind DNA forming N"-(3"-monophospho-deoxyguanosin-8-yl)-N-acetylbenzidine (dGp-ABZ). dgp-abz 141-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 12207498-0 2002 Metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) by human cytochrome P450-dependent monooxygenase systems. Polychlorinated Dibenzodioxins 14-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 12220536-0 2002 Metal-mediated DNA damage induced by curcumin in the presence of human cytochrome P450 isozymes. Metals 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 12220536-0 2002 Metal-mediated DNA damage induced by curcumin in the presence of human cytochrome P450 isozymes. Curcumin 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 12093814-2 2002 Cytochrome P450 (P450) 2D6 is involved in the oxidation of a large fraction ( approximately 30%) of drugs used by humans and also catalyzes the O-demethylation of the model substrates 3- and 4-methoxyphenethylamine followed by subsequent ring hydroxylation to dopamine. 3- and 4-methoxyphenethylamine 184-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 12093814-2 2002 Cytochrome P450 (P450) 2D6 is involved in the oxidation of a large fraction ( approximately 30%) of drugs used by humans and also catalyzes the O-demethylation of the model substrates 3- and 4-methoxyphenethylamine followed by subsequent ring hydroxylation to dopamine. Dopamine 260-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 12207498-0 2002 Metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) by human cytochrome P450-dependent monooxygenase systems. Polychlorinated Dibenzodioxins 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 12207498-1 2002 Metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) by monooxygenase systems dependent on 12 forms of human cytochrome P450 (CYP) was examined with the recombinant yeast microsomes containing each of the human CYP. Polychlorinated Dibenzodioxins 14-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-127 12206675-1 2002 Cytochrome P450 (P450) 2D6 was first identified as the polymorphic human debrisoquine hydroxylase and subsequently shown to catalyze the oxidation of a variety of drugs containing a basic nitrogen. Nitrogen 188-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 12207498-1 2002 Metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) by monooxygenase systems dependent on 12 forms of human cytochrome P450 (CYP) was examined with the recombinant yeast microsomes containing each of the human CYP. Polychlorinated Dibenzodioxins 14-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-132 12207498-1 2002 Metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) by monooxygenase systems dependent on 12 forms of human cytochrome P450 (CYP) was examined with the recombinant yeast microsomes containing each of the human CYP. Polychlorinated Dibenzodioxins 14-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-217 12207498-1 2002 Metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) by monooxygenase systems dependent on 12 forms of human cytochrome P450 (CYP) was examined with the recombinant yeast microsomes containing each of the human CYP. Polychlorinated Dibenzodioxins 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-127 12207498-1 2002 Metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) by monooxygenase systems dependent on 12 forms of human cytochrome P450 (CYP) was examined with the recombinant yeast microsomes containing each of the human CYP. Polychlorinated Dibenzodioxins 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-132 12207498-1 2002 Metabolism of polychlorinated dibenzo-p-dioxins (PCDDs) by monooxygenase systems dependent on 12 forms of human cytochrome P450 (CYP) was examined with the recombinant yeast microsomes containing each of the human CYP. Polychlorinated Dibenzodioxins 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-217 12207498-3 2002 Remarkable metabolism by the multiple CYP forms was observed toward dibenzo-p-dioxin (DD) and mono-, di-, and trichloroDDs. dibenzo(1,4)dioxin 68-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 12207498-3 2002 Remarkable metabolism by the multiple CYP forms was observed toward dibenzo-p-dioxin (DD) and mono-, di-, and trichloroDDs. dibenzo(1,4)dioxin 86-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 12207498-3 2002 Remarkable metabolism by the multiple CYP forms was observed toward dibenzo-p-dioxin (DD) and mono-, di-, and trichloroDDs. mono-, di-, and trichlorodds 94-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 12363340-2 2002 Limitation of the dissolved oxygen supplied during cultivation of various microbial strains can decrease the activity of cytochrome P-450 monooxygenases required for the processing of pathway intermediates into their final forms, resulting in the accumulation of these intermediates as the primary products. Oxygen 28-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 12236850-0 2002 Involvement of CYP2C9 and UGT2B7 in the metabolism of zaltoprofen, a nonsteroidal anti-inflammatory drug, and its lack of clinically significant CYP inhibition potential. pyranoprofen 54-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 12236850-1 2002 AIMS: To identify the cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) isoforms responsible for the formation of the primary metabolite(s) of zaltoprofen, and to predict possible drug interactions by investigating the inhibition of CYP isoforms in vitro. pyranoprofen 153-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 12236850-1 2002 AIMS: To identify the cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) isoforms responsible for the formation of the primary metabolite(s) of zaltoprofen, and to predict possible drug interactions by investigating the inhibition of CYP isoforms in vitro. pyranoprofen 153-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 12236850-1 2002 AIMS: To identify the cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) isoforms responsible for the formation of the primary metabolite(s) of zaltoprofen, and to predict possible drug interactions by investigating the inhibition of CYP isoforms in vitro. pyranoprofen 153-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 243-246 12236850-2 2002 METHODS: The metabolism of zaltoprofen was studied in vitro using recombinant CYP and UGT isoform cDNA-expression systems. pyranoprofen 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 12236850-13 2002 CONCLUSIONS: Zaltoprofen is predominantly metabolized by CYP2C9 and UGT2B7, and is considered unlikely to cause significant drug interactions in vivo when coadministered with CYP substrates at clinically effective doses. pyranoprofen 13-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 12167570-1 2002 The metabolism of methoxychlor, a proestrogenic pesticide (endocrine disruptor), was investigated with cDNA expressed human cytochrome P450s and liver microsomes (HLM). Methoxychlor 18-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 12192156-0 2002 Biomimetic oxidation of 2-methylimidazole derivative with a chemical model system for cytochrome P-450. 2-methylimidazole 24-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 12167460-1 2002 Many studies have demonstrated that cyclophosphamide (CPA) can affect hepatic cytochrome p450 (CYP) isoenzyme activity in animals. Cyclophosphamide 36-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 12167460-1 2002 Many studies have demonstrated that cyclophosphamide (CPA) can affect hepatic cytochrome p450 (CYP) isoenzyme activity in animals. Cyclophosphamide 36-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-98 12167460-1 2002 Many studies have demonstrated that cyclophosphamide (CPA) can affect hepatic cytochrome p450 (CYP) isoenzyme activity in animals. Cyclophosphamide 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 12167460-1 2002 Many studies have demonstrated that cyclophosphamide (CPA) can affect hepatic cytochrome p450 (CYP) isoenzyme activity in animals. Cyclophosphamide 54-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-98 12167460-2 2002 We have investigated the effect of CPA on gene expression of various CYP enzymes as well as beta-actin in the human acute promyelocytic leukemia cell line (HL-60S) and its multidrug-resistant (MDR) phenotype HL-60R. Cyclophosphamide 35-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 12167460-4 2002 In determination of cytotoxicity and resistance factor (RF: IC(50) HL-60R/IC(50) HL-60S), concentrations of 100 and 500 micro g/ml CPA were selected to treat HL-60S and HL-60R up to 72 h. CYP gene expression in the cells prior to and after treatment with CPA was determined using semiquantitative reverse transcriptase-polymerase chain reaction (RT-PCR) and real-time PCR. Cyclophosphamide 131-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-191 12172927-3 2002 Although CYP and GST enzymes are involved in the activation and detoxification of N-nitrosamines and related compound, studies on the relationship between genetic polymorphisms of CYP2E1, GSTT1, and GSTM1 and the risk of gastric carcinoma (GC) are few, and the results have been conflicting. n-nitrosamines 82-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-12 12192156-1 2002 A chemical model system for cytochrome P-450, consisting of tetraphenylporphyrin manganese chloride (TPPMnCl) and iodosylbenzene, efficiently oxidized 2-methylimidazole to 2-methylimidazolone. tetraphenylporphyrin manganese chloride 60-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 12192156-1 2002 A chemical model system for cytochrome P-450, consisting of tetraphenylporphyrin manganese chloride (TPPMnCl) and iodosylbenzene, efficiently oxidized 2-methylimidazole to 2-methylimidazolone. tppmncl 101-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 12192156-1 2002 A chemical model system for cytochrome P-450, consisting of tetraphenylporphyrin manganese chloride (TPPMnCl) and iodosylbenzene, efficiently oxidized 2-methylimidazole to 2-methylimidazolone. iodosobenzene 114-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 12171873-2 2002 IFNs affect cytochrome P450 (CYP) enzymes, which metabolize many endogenous (e.g., steroids, fatty acids) and exogenous (e.g., drugs) substrates. Steroids 83-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 12192156-1 2002 A chemical model system for cytochrome P-450, consisting of tetraphenylporphyrin manganese chloride (TPPMnCl) and iodosylbenzene, efficiently oxidized 2-methylimidazole to 2-methylimidazolone. 2-methylimidazole 151-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 12171873-2 2002 IFNs affect cytochrome P450 (CYP) enzymes, which metabolize many endogenous (e.g., steroids, fatty acids) and exogenous (e.g., drugs) substrates. Steroids 83-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 12171873-2 2002 IFNs affect cytochrome P450 (CYP) enzymes, which metabolize many endogenous (e.g., steroids, fatty acids) and exogenous (e.g., drugs) substrates. Fatty Acids 93-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 12192156-1 2002 A chemical model system for cytochrome P-450, consisting of tetraphenylporphyrin manganese chloride (TPPMnCl) and iodosylbenzene, efficiently oxidized 2-methylimidazole to 2-methylimidazolone. 2-methylimidazolone 172-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 12171873-2 2002 IFNs affect cytochrome P450 (CYP) enzymes, which metabolize many endogenous (e.g., steroids, fatty acids) and exogenous (e.g., drugs) substrates. Fatty Acids 93-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 12187294-2 2002 However, this could possibly influence vitamin D 25-hydroxylation because this reaction is catalyzed in part by the mitochondrial cytochrome P-450, the enzyme responsible for the 27-hydroxylation of cholesterol. Vitamin D 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 12124303-0 2002 Metabolism of tamoxifen by recombinant human cytochrome P450 enzymes: formation of the 4-hydroxy, 4"-hydroxy and N-desmethyl metabolites and isomerization of trans-4-hydroxytamoxifen. Tamoxifen 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 12124303-0 2002 Metabolism of tamoxifen by recombinant human cytochrome P450 enzymes: formation of the 4-hydroxy, 4"-hydroxy and N-desmethyl metabolites and isomerization of trans-4-hydroxytamoxifen. 4-hydroxy, 4"-hydroxy 87-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 12124303-0 2002 Metabolism of tamoxifen by recombinant human cytochrome P450 enzymes: formation of the 4-hydroxy, 4"-hydroxy and N-desmethyl metabolites and isomerization of trans-4-hydroxytamoxifen. hydroxytamoxifen 158-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 12124303-1 2002 The cytochrome P450 (P450)-mediated biotransformation of tamoxifen is important in determining both the clearance of the drug and its conversion to the active metabolite, trans-4-hydroxytamoxifen. Tamoxifen 57-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 12124303-1 2002 The cytochrome P450 (P450)-mediated biotransformation of tamoxifen is important in determining both the clearance of the drug and its conversion to the active metabolite, trans-4-hydroxytamoxifen. hydroxytamoxifen 171-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 12214661-0 2002 Electron supply and catalytic oxidation of nitrogen by cytochrome P450 and nitric oxide synthase. Nitrogen 43-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 12214661-1 2002 Cytochrome P450 and nitric oxide synthase (NOS) oxidize nitrogen atoms, although the substrates and transformations are highly restricted for NOS. Nitrogen 56-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12214668-7 2002 Preclinical studies in vivo have demonstrated that although AQ4N has little or no intrinsic cytotoxic activity per se it (i) enhances the antitumor effects of radiation and conventional chemotherapeutic agents, (ii) is pharmacokinetically stable, and (iii) is a substrate for cytochrome P450 (CYP). AQ4N 60-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 276-291 12214668-7 2002 Preclinical studies in vivo have demonstrated that although AQ4N has little or no intrinsic cytotoxic activity per se it (i) enhances the antitumor effects of radiation and conventional chemotherapeutic agents, (ii) is pharmacokinetically stable, and (iii) is a substrate for cytochrome P450 (CYP). AQ4N 60-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 293-296 12214668-8 2002 A study of AQ4N metabolism in vitro and ex vivo using purified CYP enzymes, phenotyped human livers and CYP transfected cell lines shows that CYP3A, 1A and 1B1 family members contribute to AQ4N bioreduction in the absence of oxygen. AQ4N 11-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 12214668-8 2002 A study of AQ4N metabolism in vitro and ex vivo using purified CYP enzymes, phenotyped human livers and CYP transfected cell lines shows that CYP3A, 1A and 1B1 family members contribute to AQ4N bioreduction in the absence of oxygen. AQ4N 11-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-107 12214668-8 2002 A study of AQ4N metabolism in vitro and ex vivo using purified CYP enzymes, phenotyped human livers and CYP transfected cell lines shows that CYP3A, 1A and 1B1 family members contribute to AQ4N bioreduction in the absence of oxygen. AQ4N 189-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 12214668-8 2002 A study of AQ4N metabolism in vitro and ex vivo using purified CYP enzymes, phenotyped human livers and CYP transfected cell lines shows that CYP3A, 1A and 1B1 family members contribute to AQ4N bioreduction in the absence of oxygen. Oxygen 225-231 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 12214668-9 2002 Importantly AQ4N is shown to be metabolized by tumors known to express CYP isoforms. AQ4N 12-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 12214673-0 2002 Role of human cytochrome P450 (CYP) in the metabolic activation of nitrosamine derivatives: application of genetically engineered Salmonella expressing human CYP. Nitrosamines 67-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 12214673-0 2002 Role of human cytochrome P450 (CYP) in the metabolic activation of nitrosamine derivatives: application of genetically engineered Salmonella expressing human CYP. Nitrosamines 67-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 12214673-0 2002 Role of human cytochrome P450 (CYP) in the metabolic activation of nitrosamine derivatives: application of genetically engineered Salmonella expressing human CYP. Nitrosamines 67-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-161 12201364-0 2002 Cold induces catalytic iron release of cytochrome P-450 origin: a critical step in cold storage-induced renal injury. Iron 23-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 12201364-2 2002 Whether the cytochrome P-450 enzymes, shown to be a source for iron in several injury models, contribute to cold-induced iron release is not known. Iron 63-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 12201364-2 2002 Whether the cytochrome P-450 enzymes, shown to be a source for iron in several injury models, contribute to cold-induced iron release is not known. Iron 121-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 12201364-5 2002 As microsomes are rich in iron-containing cytochrome P-450 enzymes, microsomes were cold stored with P-450 inhibitors, cimetidine and piperonyl butoxide. Iron 26-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 12201364-10 2002 Our data demonstrate for the first time that cold-induced catalytic iron release may be at least in part of microsomal cytochrome P-450 origin, and that it participates in cold-storage-induced renal injury. Iron 68-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 12187294-2 2002 However, this could possibly influence vitamin D 25-hydroxylation because this reaction is catalyzed in part by the mitochondrial cytochrome P-450, the enzyme responsible for the 27-hydroxylation of cholesterol. Cholesterol 199-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 12135382-1 2002 Cytochrome P450(BM3)-F87G reacts with aromatic aldehydes and hydrogen peroxide to generate covalent heme adducts in a reaction that may involve the formation of a stable isoporphyrin intermediate [Raner, G. M., Hatchell, A. J., Morton, P. E., Ballou, D. P., and Coon, M. J. Aldehydes 47-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-19 12135382-1 2002 Cytochrome P450(BM3)-F87G reacts with aromatic aldehydes and hydrogen peroxide to generate covalent heme adducts in a reaction that may involve the formation of a stable isoporphyrin intermediate [Raner, G. M., Hatchell, A. J., Morton, P. E., Ballou, D. P., and Coon, M. J. Hydrogen Peroxide 61-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-19 12135382-1 2002 Cytochrome P450(BM3)-F87G reacts with aromatic aldehydes and hydrogen peroxide to generate covalent heme adducts in a reaction that may involve the formation of a stable isoporphyrin intermediate [Raner, G. M., Hatchell, A. J., Morton, P. E., Ballou, D. P., and Coon, M. J. Heme 100-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-19 12135382-1 2002 Cytochrome P450(BM3)-F87G reacts with aromatic aldehydes and hydrogen peroxide to generate covalent heme adducts in a reaction that may involve the formation of a stable isoporphyrin intermediate [Raner, G. M., Hatchell, A. J., Morton, P. E., Ballou, D. P., and Coon, M. J. isoporphyrin 170-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-19 12126389-0 2002 Catalytic oxidations of steroid substrates by artificial cytochrome p-450 enzymes. Steroids 24-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 12123654-1 2002 The reaction of demethylation mediated by cytochrome P450 (CYP) leads to the equimolar production of demethylated metabolite and formaldehyde. Formaldehyde 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 12123750-0 2002 Covalent binding of the anticancer drug ellipticine to DNA in V79 cells transfected with human cytochrome P450 enzymes. ellipticine 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 12123654-1 2002 The reaction of demethylation mediated by cytochrome P450 (CYP) leads to the equimolar production of demethylated metabolite and formaldehyde. Formaldehyde 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 12123654-5 2002 The method first involves the limiting step of the CYP-dependent reaction, which is stopped with a mixture of zinc sulfate 5 mM and trichloroacetic acid 100 mM. Zinc Sulfate 110-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 12123750-2 2002 Recently, we found that ellipticine also forms covalent DNA adducts and that the formation of the major adduct is dependent on the activation of ellipticine by cytochrome P450 (CYP). ellipticine 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-175 12123654-5 2002 The method first involves the limiting step of the CYP-dependent reaction, which is stopped with a mixture of zinc sulfate 5 mM and trichloroacetic acid 100 mM. Trichloroacetic Acid 132-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 12123750-2 2002 Recently, we found that ellipticine also forms covalent DNA adducts and that the formation of the major adduct is dependent on the activation of ellipticine by cytochrome P450 (CYP). ellipticine 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 12123750-2 2002 Recently, we found that ellipticine also forms covalent DNA adducts and that the formation of the major adduct is dependent on the activation of ellipticine by cytochrome P450 (CYP). ellipticine 145-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-175 12396131-6 2002 Homology models of both the chimeric proteins were developed using SWISS-MODEL based on the known crystal structure of cytochrome P-450 camC, BM-3, 1DT6A, and 2C17A. camc 136-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 12123750-2 2002 Recently, we found that ellipticine also forms covalent DNA adducts and that the formation of the major adduct is dependent on the activation of ellipticine by cytochrome P450 (CYP). ellipticine 145-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 12123750-3 2002 We examined a panel of genetically engineered V79 cell lines including the parental line V79MZ and recombinant cells expressing the human CYP enzymes CYP1A1, CYP1A2 or CYP3A4 for their ability to activate ellipticine. ellipticine 205-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 12123750-6 2002 The nuclease P1 version of the 32P-postlabelling assay yielded a similar pattern of ellipticine-DNA adducts with two major adducts in all cells, the formation of only one of which was dependent on CYP activity. Phosphorus-32 31-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-200 12123750-10 2002 The results presented here are the first report showing the formation of CYP-mediated covalent DNA adducts by ellipticine in cells in culture, and confirm the formation of covalent DNA adducts as a new mechanism of ellipticine action. ellipticine 110-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 12123750-10 2002 The results presented here are the first report showing the formation of CYP-mediated covalent DNA adducts by ellipticine in cells in culture, and confirm the formation of covalent DNA adducts as a new mechanism of ellipticine action. ellipticine 215-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 12107550-0 2002 Cytochrome P450 isozymes 3A4 and 2B6 are involved in the in vitro human metabolism of thiotepa to TEPA. Thiotepa 86-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12084621-0 2002 Imidacloprid insecticide metabolism: human cytochrome P450 isozymes differ in selectivity for imidazolidine oxidation versus nitroimine reduction. imidacloprid 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 12084621-0 2002 Imidacloprid insecticide metabolism: human cytochrome P450 isozymes differ in selectivity for imidazolidine oxidation versus nitroimine reduction. Imidazolidines 94-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 12084621-0 2002 Imidacloprid insecticide metabolism: human cytochrome P450 isozymes differ in selectivity for imidazolidine oxidation versus nitroimine reduction. nitroimine 125-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. NADP 133-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. NADP 133-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. Alkenes 191-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. Alkenes 191-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. Imidazolidines 261-274 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. Imidazolidines 261-274 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. nitrosoimine 290-302 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. nitrosoimine 290-302 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. Guanidine 312-321 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. Guanidine 312-321 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. Urea 334-338 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. Urea 334-338 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. nitroimine 394-404 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 12084621-2 2002 This study with individual recombinant cytochrome P450 (CYP450) isozymes from human liver shows that the principal organoextractable NADPH-dependent metabolites are the 5-hydroxy (major) and olefin (minor) derivatives from hydroxylation and desaturation of the imidazolidine moiety and the nitrosoimine (major), guanidine (minor) and urea (trace) derivatives from reduction and cleavage of the nitroimine substituent. nitroimine 394-404 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-62 12087352-0 2002 Effect of interferon alpha-ribavirin bitherapy on cytochrome P450 1A2 and 2D6 and N-acetyltransferase-2 activities in patients with chronic active hepatitis C. BACKGROUND: Interferon alpha (IFN-alpha) is thought to be responsible for cytochrome P450 (CYP)-dependent drug interactions mediated by a decrease in CYP activities. alpha-Ribavirin 21-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 12060642-1 2002 PURPOSE: This research investigated the biotransformation of thalidomide by cytochrome P-450 (CYP). Thalidomide 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 12060642-1 2002 PURPOSE: This research investigated the biotransformation of thalidomide by cytochrome P-450 (CYP). Thalidomide 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-97 12060642-2 2002 EXPERIMENTAL DESIGN: We used liver microsomes from humans and/or animals and the recombinant specific CYP isozymes to investigate CYP-mediated metabolism of thalidomide. Thalidomide 157-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 12107550-0 2002 Cytochrome P450 isozymes 3A4 and 2B6 are involved in the in vitro human metabolism of thiotepa to TEPA. Triethylenephosphoramide 98-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12060642-2 2002 EXPERIMENTAL DESIGN: We used liver microsomes from humans and/or animals and the recombinant specific CYP isozymes to investigate CYP-mediated metabolism of thalidomide. Thalidomide 157-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 12107550-1 2002 PURPOSE: To establish the cytochrome P450 (CYP) isozymes involved in the metabolism of the alkylating agent, thiotepa, to the pharmacologically active metabolite, TEPA. Thiotepa 109-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 12107550-1 2002 PURPOSE: To establish the cytochrome P450 (CYP) isozymes involved in the metabolism of the alkylating agent, thiotepa, to the pharmacologically active metabolite, TEPA. Thiotepa 109-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 12107550-1 2002 PURPOSE: To establish the cytochrome P450 (CYP) isozymes involved in the metabolism of the alkylating agent, thiotepa, to the pharmacologically active metabolite, TEPA. Triethylenephosphoramide 163-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 12107550-1 2002 PURPOSE: To establish the cytochrome P450 (CYP) isozymes involved in the metabolism of the alkylating agent, thiotepa, to the pharmacologically active metabolite, TEPA. Triethylenephosphoramide 163-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 12107550-10 2002 There is a potential for CYP-mediated drug interactions with thiotepa. Thiotepa 61-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 12107550-11 2002 Pharmacokinetic variability of thiotepa may be related to expression of hepatic CYP isozymes. Thiotepa 31-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 12027667-4 2002 The reagent was exploited in a total synthesis of 13-oxotridec-9E,11E-dienoic acid, confirming the identity of this product that is generated upon autoxidation of linoleic acid and by decomposition of 13-hydroperoxy-9,11-octadecadienoate (13-HPODE), especially in the presence of redox active transition metal ions, cytochrome p-450, or hydroperoxide lyase. 13-oxotridec-9e 50-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 316-332 12020594-0 2002 Comparative effect of colchicine and colchiceine on cytotoxicity and CYP gene expression in primary human hepatocytes. colchiceine 37-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 12027667-4 2002 The reagent was exploited in a total synthesis of 13-oxotridec-9E,11E-dienoic acid, confirming the identity of this product that is generated upon autoxidation of linoleic acid and by decomposition of 13-hydroperoxy-9,11-octadecadienoate (13-HPODE), especially in the presence of redox active transition metal ions, cytochrome p-450, or hydroperoxide lyase. 11e-dienoic acid 66-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 316-332 12027667-4 2002 The reagent was exploited in a total synthesis of 13-oxotridec-9E,11E-dienoic acid, confirming the identity of this product that is generated upon autoxidation of linoleic acid and by decomposition of 13-hydroperoxy-9,11-octadecadienoate (13-HPODE), especially in the presence of redox active transition metal ions, cytochrome p-450, or hydroperoxide lyase. Linoleic Acid 163-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 316-332 12027667-4 2002 The reagent was exploited in a total synthesis of 13-oxotridec-9E,11E-dienoic acid, confirming the identity of this product that is generated upon autoxidation of linoleic acid and by decomposition of 13-hydroperoxy-9,11-octadecadienoate (13-HPODE), especially in the presence of redox active transition metal ions, cytochrome p-450, or hydroperoxide lyase. 13-hydroperoxy-9,11-octadecadienoate 201-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 316-332 12027667-4 2002 The reagent was exploited in a total synthesis of 13-oxotridec-9E,11E-dienoic acid, confirming the identity of this product that is generated upon autoxidation of linoleic acid and by decomposition of 13-hydroperoxy-9,11-octadecadienoate (13-HPODE), especially in the presence of redox active transition metal ions, cytochrome p-450, or hydroperoxide lyase. Metals 304-309 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 316-332 12033517-0 2002 Inhibition of human drug metabolizing cytochrome P450 by buprenorphine. Buprenorphine 57-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 12076522-0 2002 Kinetics of bromodichloromethane metabolism by cytochrome P450 isoenzymes in human liver microsomes. bromodichloromethane 12-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 12076522-1 2002 The kinetic constants for the metabolism of bromodichloromethane (BDCM) by three cytochrome P450 (CYP) isoenzymes have been measured in human liver microsomes. bromodichloromethane 44-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 12076522-1 2002 The kinetic constants for the metabolism of bromodichloromethane (BDCM) by three cytochrome P450 (CYP) isoenzymes have been measured in human liver microsomes. bromodichloromethane 44-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 12076522-1 2002 The kinetic constants for the metabolism of bromodichloromethane (BDCM) by three cytochrome P450 (CYP) isoenzymes have been measured in human liver microsomes. bromodichloromethane 66-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 12076522-1 2002 The kinetic constants for the metabolism of bromodichloromethane (BDCM) by three cytochrome P450 (CYP) isoenzymes have been measured in human liver microsomes. bromodichloromethane 66-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 12076522-12 2002 Because of the low levels of BDCM exposure from drinking water, it appears likely that CYP2E1 will dominate hepatic CYP-mediated BDCM metabolism in humans. Water 57-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 12076522-12 2002 Because of the low levels of BDCM exposure from drinking water, it appears likely that CYP2E1 will dominate hepatic CYP-mediated BDCM metabolism in humans. bromodichloromethane 129-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 12033517-1 2002 The effects of buprenorphine, a powerful mixed agonist/antagonist analgesic, on several cytochrome P450 (CYP) isoform specific reactions in human liver microsomes were investigated to predict drug interaction of buprenorphine in vivo from in vitro data. Buprenorphine 15-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 12033517-1 2002 The effects of buprenorphine, a powerful mixed agonist/antagonist analgesic, on several cytochrome P450 (CYP) isoform specific reactions in human liver microsomes were investigated to predict drug interaction of buprenorphine in vivo from in vitro data. Buprenorphine 15-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-108 12033517-1 2002 The effects of buprenorphine, a powerful mixed agonist/antagonist analgesic, on several cytochrome P450 (CYP) isoform specific reactions in human liver microsomes were investigated to predict drug interaction of buprenorphine in vivo from in vitro data. Buprenorphine 212-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-108 11950794-4 2002 Sulfaphenazole, flavoxamine, and antibodies raised against purified liver cytochrome P450 (P450) 2C9 that inhibit both CYP2C9- and 2C19-dependent activities, significantly inhibited microsomal oxidations of (+)- and (-)-limonene enantiomers. flavoxamine 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-100 12067002-9 2002 RESULTS: Total recovery of propofol in the metabolites studied amounts to 38%, of which 62% was via the PG metabolite and 38% via cytochrome P-450. Propofol 27-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 11950794-4 2002 Sulfaphenazole, flavoxamine, and antibodies raised against purified liver cytochrome P450 (P450) 2C9 that inhibit both CYP2C9- and 2C19-dependent activities, significantly inhibited microsomal oxidations of (+)- and (-)-limonene enantiomers. (+)- and (-)-limonene 207-228 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-100 11996890-9 2002 Since these cells lack the isoenzymatic form of cytochrome P(450) mainly involved in the ethanol metabolism (namely cytochrome P(450)2E1) and also are devoid of alcohol dehydrogenase activity, we propose that the toxic actions of ethanol on liver must be linked to the activity of one or both of these systems. Ethanol 89-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-65 11964369-2 2002 Following computer-based molecule similarity, we proposed that on cytochrome-P450 degradation, the LOS metabolite EXP3179 is generated, which shows molecule homology to indomethacin, a cyclooxygenase inhibitor with antiinflammatory and antiaggregatory properties. Indomethacin 169-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 12422544-18 2002 Another theory involves toxic metabolites; the aromatic antiepileptic agents are metabolised by cytochrome P-450 to an arene oxide metabolite. Benzene oxide 119-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 11996890-9 2002 Since these cells lack the isoenzymatic form of cytochrome P(450) mainly involved in the ethanol metabolism (namely cytochrome P(450)2E1) and also are devoid of alcohol dehydrogenase activity, we propose that the toxic actions of ethanol on liver must be linked to the activity of one or both of these systems. Ethanol 230-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-65 11872324-2 2002 The current study was conducted to assess the potential for nefazodone to have metabolic drug interactions associated with cytochrome P450 (CYP) enzymes. nefazodone 60-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-138 11956503-4 2002 CYP enzyme activities were measured by means of the metabolic ratios of sparteine (CYP2D6), endogenous cortisol metabolism (CYP3A4), and caffeine (CYP1A2), as well as by the S/R ratio of mephenytoin (CYP2C19) and antipyrine clearance. Sparteine 72-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 11956503-4 2002 CYP enzyme activities were measured by means of the metabolic ratios of sparteine (CYP2D6), endogenous cortisol metabolism (CYP3A4), and caffeine (CYP1A2), as well as by the S/R ratio of mephenytoin (CYP2C19) and antipyrine clearance. Hydrocortisone 103-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 11956503-4 2002 CYP enzyme activities were measured by means of the metabolic ratios of sparteine (CYP2D6), endogenous cortisol metabolism (CYP3A4), and caffeine (CYP1A2), as well as by the S/R ratio of mephenytoin (CYP2C19) and antipyrine clearance. Caffeine 137-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 11956503-4 2002 CYP enzyme activities were measured by means of the metabolic ratios of sparteine (CYP2D6), endogenous cortisol metabolism (CYP3A4), and caffeine (CYP1A2), as well as by the S/R ratio of mephenytoin (CYP2C19) and antipyrine clearance. Mephenytoin 187-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 11956503-4 2002 CYP enzyme activities were measured by means of the metabolic ratios of sparteine (CYP2D6), endogenous cortisol metabolism (CYP3A4), and caffeine (CYP1A2), as well as by the S/R ratio of mephenytoin (CYP2C19) and antipyrine clearance. Antipyrine 213-223 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 11956677-1 2002 OBJECTIVE: To determine whether the antiprotozoal drug atovaquone inhibits the cytochrome P(450) (CYP)2C9-mediated metabolism of sulphamethoxazole (SMX) to its potentially harmful hydroxylamine metabolite (SMX-HA) in vitro. Atovaquone 55-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-96 11872324-2 2002 The current study was conducted to assess the potential for nefazodone to have metabolic drug interactions associated with cytochrome P450 (CYP) enzymes. nefazodone 60-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-143 11956677-1 2002 OBJECTIVE: To determine whether the antiprotozoal drug atovaquone inhibits the cytochrome P(450) (CYP)2C9-mediated metabolism of sulphamethoxazole (SMX) to its potentially harmful hydroxylamine metabolite (SMX-HA) in vitro. Sulfamethoxazole 129-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-96 11956677-1 2002 OBJECTIVE: To determine whether the antiprotozoal drug atovaquone inhibits the cytochrome P(450) (CYP)2C9-mediated metabolism of sulphamethoxazole (SMX) to its potentially harmful hydroxylamine metabolite (SMX-HA) in vitro. Sulfamethoxazole 148-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-96 12028321-11 2002 Frovatriptan is principally metabolized by the CYP1A2 isoenzyme of cytochrome P-450 and is cleared by the kidney and liver, each having sufficient capacity to compensate for impairment of the other. frovatriptan 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 11866476-2 2002 Cytochrome P450-dependent oxidation of acetaminophen results in the formation of the toxic N-acetyl-p-benzoquinone-imine (NAPQI). Acetaminophen 39-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11891538-0 2002 The environmental toxin 2,3,7,8-tetrachlorodibenzo-p-dioxin induces cytochrome P450 activity in high passage PC 3 and DU 145 human prostate cancer cell lines. Polychlorinated Dibenzodioxins 24-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 11891538-0 2002 The environmental toxin 2,3,7,8-tetrachlorodibenzo-p-dioxin induces cytochrome P450 activity in high passage PC 3 and DU 145 human prostate cancer cell lines. du 118-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 11891538-6 2002 The induction of CYP1A1 and CYP1B1 by TCCD in the hormone-independent prostate cancer cell lines suggests that CYP induction should be considered in patients with advanced prostate cancer. tccd 38-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 11983405-1 2002 An overview of the application of high-pressure studies on the carbon monoxide complex of cytochrome P-450 is given. Carbon Monoxide 63-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 11967242-5 2002 The inhibitory effects of estradiol on VSMCs were enhanced by cytochrome-P450 (CYP450) inducers 3-methylcholanthrene and phenobarbital. Estradiol 26-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 11967242-5 2002 The inhibitory effects of estradiol on VSMCs were enhanced by cytochrome-P450 (CYP450) inducers 3-methylcholanthrene and phenobarbital. Estradiol 26-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-85 11967242-5 2002 The inhibitory effects of estradiol on VSMCs were enhanced by cytochrome-P450 (CYP450) inducers 3-methylcholanthrene and phenobarbital. vsmcs 39-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 11967242-5 2002 The inhibitory effects of estradiol on VSMCs were enhanced by cytochrome-P450 (CYP450) inducers 3-methylcholanthrene and phenobarbital. vsmcs 39-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-85 11967242-5 2002 The inhibitory effects of estradiol on VSMCs were enhanced by cytochrome-P450 (CYP450) inducers 3-methylcholanthrene and phenobarbital. Methylcholanthrene 96-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 11967242-5 2002 The inhibitory effects of estradiol on VSMCs were enhanced by cytochrome-P450 (CYP450) inducers 3-methylcholanthrene and phenobarbital. Methylcholanthrene 96-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-85 11967242-5 2002 The inhibitory effects of estradiol on VSMCs were enhanced by cytochrome-P450 (CYP450) inducers 3-methylcholanthrene and phenobarbital. Phenobarbital 121-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 11967242-5 2002 The inhibitory effects of estradiol on VSMCs were enhanced by cytochrome-P450 (CYP450) inducers 3-methylcholanthrene and phenobarbital. Phenobarbital 121-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-85 11967242-6 2002 Moreover, the inhibitory effects of estradiol were blocked in the presence of the CYP450 inhibitor 1-aminobenzotriazole and the catechol-O-methyltransferase inhibitors quercetin and OR486. Estradiol 36-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-88 11967242-6 2002 Moreover, the inhibitory effects of estradiol were blocked in the presence of the CYP450 inhibitor 1-aminobenzotriazole and the catechol-O-methyltransferase inhibitors quercetin and OR486. 1-aminobenzotriazole 99-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-88 11869873-2 2002 The environmental pollutant 3-methylsulfonyl-DDE (3-MeSO2-DDE) and the cytochrome P450 (CYP) inhibitors ketoconazole, metyrapone and aminoglutethimide were studied for their effects on the steroid formation. Ketoconazole 104-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 11869873-2 2002 The environmental pollutant 3-methylsulfonyl-DDE (3-MeSO2-DDE) and the cytochrome P450 (CYP) inhibitors ketoconazole, metyrapone and aminoglutethimide were studied for their effects on the steroid formation. Metyrapone 118-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 11869873-2 2002 The environmental pollutant 3-methylsulfonyl-DDE (3-MeSO2-DDE) and the cytochrome P450 (CYP) inhibitors ketoconazole, metyrapone and aminoglutethimide were studied for their effects on the steroid formation. Aminoglutethimide 133-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 11869873-2 2002 The environmental pollutant 3-methylsulfonyl-DDE (3-MeSO2-DDE) and the cytochrome P450 (CYP) inhibitors ketoconazole, metyrapone and aminoglutethimide were studied for their effects on the steroid formation. Steroids 189-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 11866476-2 2002 Cytochrome P450-dependent oxidation of acetaminophen results in the formation of the toxic N-acetyl-p-benzoquinone-imine (NAPQI). N-acetyl-4-benzoquinoneimine 91-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11866476-2 2002 Cytochrome P450-dependent oxidation of acetaminophen results in the formation of the toxic N-acetyl-p-benzoquinone-imine (NAPQI). N-acetyl-4-benzoquinoneimine 122-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11866476-3 2002 Inhibition of cytochrome P450 enzymes responsible for NAPQI formation might be useful--besides N-acetylcysteine treatment--in managing acetaminophen overdose. N-acetyl-4-benzoquinoneimine 54-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 11866476-3 2002 Inhibition of cytochrome P450 enzymes responsible for NAPQI formation might be useful--besides N-acetylcysteine treatment--in managing acetaminophen overdose. Acetylcysteine 95-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 11866476-3 2002 Inhibition of cytochrome P450 enzymes responsible for NAPQI formation might be useful--besides N-acetylcysteine treatment--in managing acetaminophen overdose. Acetaminophen 135-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 11866476-4 2002 Investigations were carried out using human liver microsomes to test whether selective inhibition of cytochrome P450s reduces NAPQI formation. N-acetyl-4-benzoquinoneimine 126-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 11866476-8 2002 Although cimetidin is used in the therapy of acetaminophen overdose as an inhibitor of cytochrome P450, it is not able to reduce NAPQI formation. Cimetidine 9-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 11865972-3 2002 The objective of this study is to evaluate the effect of amiodarone on the pharmacokinetics of mexiletine through its inhibition of various cytochrome P450 (CYP) subtypes. Amiodarone 57-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-155 11893129-3 2002 To better understand the interaction between genotype and response to warfarin therapy, we determined the frequency and functional effects of polymorphisms of the predominant cytochrome P450 subfamily responsible for warfarin metabolism (eg, CYP2C9) in an ethnically defined U.S. patient population. Warfarin 70-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-190 11893129-3 2002 To better understand the interaction between genotype and response to warfarin therapy, we determined the frequency and functional effects of polymorphisms of the predominant cytochrome P450 subfamily responsible for warfarin metabolism (eg, CYP2C9) in an ethnically defined U.S. patient population. Warfarin 217-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-190 11893129-11 2002 Patients with CYP polymorphisms (2C9*2, 2C9*3) had significantly lower warfarin doses compared to patients with wild-type genotypes [30.6 (+/- 2.5) mg versus 40.1 (+/- 1.7) mg, p = 0.0021] . Warfarin 71-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 11893129-12 2002 Stepwise backward regression analysis suggested a moderate ability to predict warfarin dose based on CYP genotype (r2 = 0.26), p < 0.01). Warfarin 78-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 11893129-14 2002 As interactions between genetic factors and other variables that influence warfarin effect are more completely understood, CYP analysis may prove a useful adjunct for increasing the safety and efficacy of this agent. Warfarin 75-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 11865972-3 2002 The objective of this study is to evaluate the effect of amiodarone on the pharmacokinetics of mexiletine through its inhibition of various cytochrome P450 (CYP) subtypes. Amiodarone 57-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 11865972-3 2002 The objective of this study is to evaluate the effect of amiodarone on the pharmacokinetics of mexiletine through its inhibition of various cytochrome P450 (CYP) subtypes. Mexiletine 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-155 11865972-3 2002 The objective of this study is to evaluate the effect of amiodarone on the pharmacokinetics of mexiletine through its inhibition of various cytochrome P450 (CYP) subtypes. Mexiletine 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-160 11936218-0 2002 Effects of flavonoids isolated from Scutellariae radix on cytochrome P-450 activities in human liver microsomes. Flavonoids 11-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 11936218-1 2002 A series of flavonoids isolated from Scutellariae radix were evaluated for their effects on cytochrome P-450 (CYP) activities in human liver microsomes. Flavonoids 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 11936218-1 2002 A series of flavonoids isolated from Scutellariae radix were evaluated for their effects on cytochrome P-450 (CYP) activities in human liver microsomes. Flavonoids 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 12071336-2 2002 Thus, we studied formation of the major maprotiline metabolite desmethylmaprotiline to identify the human cytochrome P-450 enzymes (CYP) involved. desmethylmaprotiline 63-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. albendazole sulfoxide 101-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 11851418-2 2002 We demonstrate here that cytochrome P450 (CYP) is responsible for the conversion of the cyano group of pinacidil to the corresponding amide. Pinacidil 103-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 11851418-2 2002 We demonstrate here that cytochrome P450 (CYP) is responsible for the conversion of the cyano group of pinacidil to the corresponding amide. Pinacidil 103-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 11851418-2 2002 We demonstrate here that cytochrome P450 (CYP) is responsible for the conversion of the cyano group of pinacidil to the corresponding amide. Amides 134-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 11851418-2 2002 We demonstrate here that cytochrome P450 (CYP) is responsible for the conversion of the cyano group of pinacidil to the corresponding amide. Amides 134-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 11851418-4 2002 Incubations of pinacidil with recombinant CYP enzymes confirm that CYP3A4 is the principal catalyst of this reaction. Pinacidil 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-45 11816012-7 2002 Since celecoxib should be metabolized primarily by cytochrome 2C9 (CYP2C9), a poor metabolizer (PM) for this cytochrome P450 enzyme was included in the study. Celecoxib 6-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 11839370-5 2002 We therefore hypothesize that the addition of lovastatin caused an increase in plasma quetiapine levels through competitive inhibition of the cytochrome P(450) (CYP) isoenzyme 3A4. Lovastatin 46-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-159 11839370-5 2002 We therefore hypothesize that the addition of lovastatin caused an increase in plasma quetiapine levels through competitive inhibition of the cytochrome P(450) (CYP) isoenzyme 3A4. Lovastatin 46-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 11839370-5 2002 We therefore hypothesize that the addition of lovastatin caused an increase in plasma quetiapine levels through competitive inhibition of the cytochrome P(450) (CYP) isoenzyme 3A4. Quetiapine Fumarate 86-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-159 11839370-5 2002 We therefore hypothesize that the addition of lovastatin caused an increase in plasma quetiapine levels through competitive inhibition of the cytochrome P(450) (CYP) isoenzyme 3A4. Quetiapine Fumarate 86-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 11847531-0 2002 Albendazole metabolism in patients with neurocysticercosis: antipyrine as a multifunctional marker drug of cytochrome P450. Albendazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 11847531-0 2002 Albendazole metabolism in patients with neurocysticercosis: antipyrine as a multifunctional marker drug of cytochrome P450. Antipyrine 60-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. albendazole sulfoxide 124-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. albendazole sulfoxide 101-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. albendazole sulfoxide 124-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. albendazole sulfone 133-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. albendazole sulfone 133-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. albendazole sulfone 154-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. albendazole sulfone 154-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. Antipyrine 202-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 11847531-1 2002 The present study investigates the isoform(s) of cytochrome P450 (CYP) involved in the metabolism of albendazole sulfoxide (ASOX) to albendazole sulfone (ASON) in patients with neurocysticercosis using antipyrine as a multifunctional marker drug. Antipyrine 202-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 11847531-9 2002 Since the CL(T) of antipyrine is a general measure of CYP enzymes but with a slight to moderate weight toward CYP1A2, we suggest the involvement of this enzyme in ASOX to ASON metabolism in man. Antipyrine 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 11847531-9 2002 Since the CL(T) of antipyrine is a general measure of CYP enzymes but with a slight to moderate weight toward CYP1A2, we suggest the involvement of this enzyme in ASOX to ASON metabolism in man. albendazole sulfone 171-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 12026186-4 2002 L-Trp also had much higher inhibitory activity on the cytochrome P-450-dependent lipid peroxidation than the previously identified antioxidants of HPE, L-phenylalanine, L-tyrosine and uracil. Tryptophan 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 11805199-0 2002 The fallacy of using adrenochrome reaction for measurement of reactive oxygen species formed during cytochrome p450-mediated metabolism of xenobiotics. Reactive Oxygen Species 62-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 11805199-1 2002 The adrenochrome reaction (oxidation of epinephrine to adrenochrome) has been widely employed as a standard assay for reactive oxygen species, produced under a variety of conditions, including those produced during cytochrome P450 (CYP)-mediated oxidation of substrates such as cyclosporine. Epinephrine 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-230 11805199-1 2002 The adrenochrome reaction (oxidation of epinephrine to adrenochrome) has been widely employed as a standard assay for reactive oxygen species, produced under a variety of conditions, including those produced during cytochrome P450 (CYP)-mediated oxidation of substrates such as cyclosporine. Epinephrine 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-235 11805199-1 2002 The adrenochrome reaction (oxidation of epinephrine to adrenochrome) has been widely employed as a standard assay for reactive oxygen species, produced under a variety of conditions, including those produced during cytochrome P450 (CYP)-mediated oxidation of substrates such as cyclosporine. Reactive Oxygen Species 118-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-230 11805199-1 2002 The adrenochrome reaction (oxidation of epinephrine to adrenochrome) has been widely employed as a standard assay for reactive oxygen species, produced under a variety of conditions, including those produced during cytochrome P450 (CYP)-mediated oxidation of substrates such as cyclosporine. Reactive Oxygen Species 118-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-235 11805199-1 2002 The adrenochrome reaction (oxidation of epinephrine to adrenochrome) has been widely employed as a standard assay for reactive oxygen species, produced under a variety of conditions, including those produced during cytochrome P450 (CYP)-mediated oxidation of substrates such as cyclosporine. Cyclosporine 278-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 215-230 11805199-1 2002 The adrenochrome reaction (oxidation of epinephrine to adrenochrome) has been widely employed as a standard assay for reactive oxygen species, produced under a variety of conditions, including those produced during cytochrome P450 (CYP)-mediated oxidation of substrates such as cyclosporine. Cyclosporine 278-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 232-235 11876575-8 2002 Sertraline is a moderate CYP2D6 inhibitor; citalopram appears to have little effect on the major CYP isoforms. Sertraline 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 11876575-9 2002 Fluoxetine deserves special attention as inhibitory effects on CYP-activity can persist for several weeks after fluoxetine discontinuation because of the long half-life of fluoxetine and its metabolite norfluoxetine. Fluoxetine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 11876575-9 2002 Fluoxetine deserves special attention as inhibitory effects on CYP-activity can persist for several weeks after fluoxetine discontinuation because of the long half-life of fluoxetine and its metabolite norfluoxetine. Fluoxetine 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 11876575-9 2002 Fluoxetine deserves special attention as inhibitory effects on CYP-activity can persist for several weeks after fluoxetine discontinuation because of the long half-life of fluoxetine and its metabolite norfluoxetine. Fluoxetine 172-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 11876575-9 2002 Fluoxetine deserves special attention as inhibitory effects on CYP-activity can persist for several weeks after fluoxetine discontinuation because of the long half-life of fluoxetine and its metabolite norfluoxetine. norfluoxetine 202-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 11805199-3 2002 Thus, in the present report, we provide evidence that measurement of adrenochrome cannot be used as an index of reactive oxygen species generated during CYP-mediated metabolism of xenobiotics because adrenochrome and its precursor, epinephrine, interact with the CYP enzyme system as substrates and inhibitors. Epinephrine 232-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-156 11805199-3 2002 Thus, in the present report, we provide evidence that measurement of adrenochrome cannot be used as an index of reactive oxygen species generated during CYP-mediated metabolism of xenobiotics because adrenochrome and its precursor, epinephrine, interact with the CYP enzyme system as substrates and inhibitors. Epinephrine 232-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 263-266 11805199-4 2002 Our results indicated that adrenochrome was moderately stable in phosphate buffer but degraded rapidly (over 50% consumed in less than 2 min) by (cloned and expressed) CYP3A4 and CYP reductase in the presence of NADPH. NADP 212-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-171 12481202-2 2002 Cytochrome p450 (CYP) metabolism of rosuvastatin appears to be minimal and is principally mediated by the 2C9 enzyme, with little involvement of 3A4; this finding is consistent with the absence of clinically significant pharmacokinetic drug-drug interactions between rosuvastatin and other drugs known to inhibit CYP enzymes. Rosuvastatin Calcium 36-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12481202-2 2002 Cytochrome p450 (CYP) metabolism of rosuvastatin appears to be minimal and is principally mediated by the 2C9 enzyme, with little involvement of 3A4; this finding is consistent with the absence of clinically significant pharmacokinetic drug-drug interactions between rosuvastatin and other drugs known to inhibit CYP enzymes. Rosuvastatin Calcium 267-279 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 12481202-2 2002 Cytochrome p450 (CYP) metabolism of rosuvastatin appears to be minimal and is principally mediated by the 2C9 enzyme, with little involvement of 3A4; this finding is consistent with the absence of clinically significant pharmacokinetic drug-drug interactions between rosuvastatin and other drugs known to inhibit CYP enzymes. Rosuvastatin Calcium 36-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 12481202-2 2002 Cytochrome p450 (CYP) metabolism of rosuvastatin appears to be minimal and is principally mediated by the 2C9 enzyme, with little involvement of 3A4; this finding is consistent with the absence of clinically significant pharmacokinetic drug-drug interactions between rosuvastatin and other drugs known to inhibit CYP enzymes. Rosuvastatin Calcium 36-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 313-316 11744607-0 2002 Induction of multidrug resistance-1 and cytochrome P450 mRNAs in human mononuclear cells by rifampin. Rifampin 92-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 12052216-1 2002 Several commonly used cancer chemotherapeutic prodrugs, including cyclophosphamide and ifosfamide, are metabolized in the liver by a cytochrome P450 (CYP)-catalyzed prodrug activation reaction that is required for therapeutic activity. Cyclophosphamide 66-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 12052216-1 2002 Several commonly used cancer chemotherapeutic prodrugs, including cyclophosphamide and ifosfamide, are metabolized in the liver by a cytochrome P450 (CYP)-catalyzed prodrug activation reaction that is required for therapeutic activity. Cyclophosphamide 66-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 12052216-1 2002 Several commonly used cancer chemotherapeutic prodrugs, including cyclophosphamide and ifosfamide, are metabolized in the liver by a cytochrome P450 (CYP)-catalyzed prodrug activation reaction that is required for therapeutic activity. Ifosfamide 87-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 12052216-1 2002 Several commonly used cancer chemotherapeutic prodrugs, including cyclophosphamide and ifosfamide, are metabolized in the liver by a cytochrome P450 (CYP)-catalyzed prodrug activation reaction that is required for therapeutic activity. Ifosfamide 87-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 12383041-1 2002 OBJECTIVE: To investigate the in vitro metabolism of the antithrombotic agent fondaparinux sodium in mammalian liver fractions and to evaluate its potential inhibitory effect on human cytochrome P450 (CYP)-mediated metabolism of other drugs. Fondaparinux 78-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 184-199 12383041-1 2002 OBJECTIVE: To investigate the in vitro metabolism of the antithrombotic agent fondaparinux sodium in mammalian liver fractions and to evaluate its potential inhibitory effect on human cytochrome P450 (CYP)-mediated metabolism of other drugs. Fondaparinux 78-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-204 12383041-5 2002 Experiments were designed to determine apparent K(i) (inhibitory constant) values for fondaparinux sodium against each CYP isoform, by varying concentrations of fondaparinux sodium and the selective substrate. Fondaparinux 86-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-122 12383041-10 2002 Apparent K(i) values for fondaparinux sodium against the CYP isoforms could not be determined because the oxidative metabolism of the isoform-selective CYP substrates was not significantly inhibited in pooled microsomal reaction mixtures. Fondaparinux 25-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 11744605-0 2002 Computational models for cytochrome P450: a predictive electronic model for aromatic oxidation and hydrogen atom abstraction. Hydrogen 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 15618670-1 2002 The participation of cytochrome P-450 (CYP) isoforms in the metabolism of selegiline was investigated. Selegiline 74-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 15618670-1 2002 The participation of cytochrome P-450 (CYP) isoforms in the metabolism of selegiline was investigated. Selegiline 74-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 15618670-2 2002 Experiments using recombinant CYP isoforms expressed in human lymphoblastoid cells showed CYP2B6 to be the major CYP isoform involved with the metabolism of selegiline. Selegiline 157-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 15618670-2 2002 Experiments using recombinant CYP isoforms expressed in human lymphoblastoid cells showed CYP2B6 to be the major CYP isoform involved with the metabolism of selegiline. Selegiline 157-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-93 11752129-4 2002 We then examined the inhibitory effects of anti-CYP4F antibody and 17-octadecynoic acid, an inhibitor of the CYP4 family, on ebastine hydroxylation in intestinal microsomes, since CYP4F was recently found to be the predominant ebastine hydroxylase in monkey intestine; and a novel CYP4F isoform (CYP4F12), also capable of hydroxylating ebastine, was found to exist in human intestine. ebastine 125-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-185 12495367-12 2002 The hepatic metabolism of rabeprazole is predominantly by nonenzymatic reactions and minimally by CYP-mediated reactions, which therefore confers an advantage over older PPIs in that genetic polymorphisms for CYP2C19 do not significantly influence rabeprazole clearance, clinical efficacy or potential for drug interactions. Rabeprazole 26-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 12060006-5 2002 A CYP-catalyzed arene oxide intermediate of LTG has been identified. Benzene oxide 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 2-5 11752129-4 2002 We then examined the inhibitory effects of anti-CYP4F antibody and 17-octadecynoic acid, an inhibitor of the CYP4 family, on ebastine hydroxylation in intestinal microsomes, since CYP4F was recently found to be the predominant ebastine hydroxylase in monkey intestine; and a novel CYP4F isoform (CYP4F12), also capable of hydroxylating ebastine, was found to exist in human intestine. ebastine 125-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-185 11752129-5 2002 However, the inhibitory effects were only partial (about 20%) and thus it was thought that, although human CYP4F was involved in ebastine hydroxylation, another predominant enzyme exists. ebastine 129-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-112 11701230-0 2001 Cytochrome P450 isozymes involved in the metabolism of phenol, a benzene metabolite. Phenol 55-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. Arachidonic Acid 35-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. Arachidonic Acid 35-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 169-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 169-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 202-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 202-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. epoxyeicosatrienoic acids 215-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. epoxyeicosatrienoic acids 215-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. eets 242-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 11773611-1 2002 Recent studies have indicated that arachidonic acid is primarily metabolized by cytochrome P-450 (CYP) enzymes in the brain, lung, kidney, and peripheral vasculature to 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) and that these compounds play critical roles in the regulation of renal, pulmonary, and cardiac function and vascular tone. eets 242-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 11773611-12 2002 Given the importance of this pathway in the control of cardiovascular function, it is likely that CYP metabolites of arachidonic acid contribute to the changes in renal function and vascular tone associated with some of these conditions and that drugs that modify the formation and/or actions of EETs and 20-HETE may have therapeutic benefits. Arachidonic Acid 117-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-101 12048797-1 2002 Patients with cirrhosis of the liver were found to have a considerable suppression of the system of biotransformation of the liver before operation which correlated with the data of the direct indices of monooxigenase system of hepatocytes--cytochrome P-450 and activity of N-demethylase of amidopyrine. Aminopyrine 291-302 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 241-287 12664659-5 2002 The CYP gene products such as CYP3A, CYP2B and PPAR are essential for metabolism of endogenous steroid hormones, fatty acids and various xenobiotics including drugs. Steroids 95-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 12664659-5 2002 The CYP gene products such as CYP3A, CYP2B and PPAR are essential for metabolism of endogenous steroid hormones, fatty acids and various xenobiotics including drugs. Fatty Acids 113-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 11701230-0 2001 Cytochrome P450 isozymes involved in the metabolism of phenol, a benzene metabolite. Benzene 65-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11701230-3 2001 To exert its toxic effects, benzene must be metabolized by cytochrome P450 to phenol and subsequently to catechol and hydroquinone. Benzene 28-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 11701230-3 2001 To exert its toxic effects, benzene must be metabolized by cytochrome P450 to phenol and subsequently to catechol and hydroquinone. Phenol 78-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 11701230-3 2001 To exert its toxic effects, benzene must be metabolized by cytochrome P450 to phenol and subsequently to catechol and hydroquinone. catechol 105-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 11829200-2 2001 METHODS: Human liver microsomes or recombinant cytochrome P450 (CYP) enzymes were incubated with either (+/- )-, (+)-, or (-)-lansoprazole in the presence of reduced nicotinamide adenine dinucleotide phosphate. (+/- )-, (+)-, or (-)-lansoprazole 104-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 11668070-1 2001 This study was conducted to test the hypothesis that the cytochrome P-450 (CYP450) metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) contributes to the afferent arteriolar response to P2 receptor activation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 94-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 11829200-2 2001 METHODS: Human liver microsomes or recombinant cytochrome P450 (CYP) enzymes were incubated with either (+/- )-, (+)-, or (-)-lansoprazole in the presence of reduced nicotinamide adenine dinucleotide phosphate. (+/- )-, (+)-, or (-)-lansoprazole 104-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 11733614-0 2001 Cytochrome P450 metabolites of arachidonic acid: novel regulators of renal function. Arachidonic Acid 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11717173-0 2001 Cytochrome P450 Involvement in the biotransformation of cisapride and racemic norcisapride in vitro: differential activity of individual human CYP3A isoforms. Cisapride 56-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11717173-0 2001 Cytochrome P450 Involvement in the biotransformation of cisapride and racemic norcisapride in vitro: differential activity of individual human CYP3A isoforms. norcisapride 78-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11717173-1 2001 Identification of the human cytochrome P450 (P450) enzymes involved in the metabolism of cisapride and racemic norcisapride [(+/-)-norcisapride] was investigated at 0.1 and 1 microM, concentrations that span the mean plasma C(max) for cisapride. Cisapride 89-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 11717173-1 2001 Identification of the human cytochrome P450 (P450) enzymes involved in the metabolism of cisapride and racemic norcisapride [(+/-)-norcisapride] was investigated at 0.1 and 1 microM, concentrations that span the mean plasma C(max) for cisapride. norcisapride 111-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 11717173-1 2001 Identification of the human cytochrome P450 (P450) enzymes involved in the metabolism of cisapride and racemic norcisapride [(+/-)-norcisapride] was investigated at 0.1 and 1 microM, concentrations that span the mean plasma C(max) for cisapride. norcisapride 125-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 11717173-1 2001 Identification of the human cytochrome P450 (P450) enzymes involved in the metabolism of cisapride and racemic norcisapride [(+/-)-norcisapride] was investigated at 0.1 and 1 microM, concentrations that span the mean plasma C(max) for cisapride. Cisapride 114-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 11714275-1 2001 Cytochrome P450 (P450) 2D6 oxidizes a wide variety of drugs typically at a distance of 5-7 A from a basic nitrogen on the substrate. Nitrogen 106-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 11668070-1 2001 This study was conducted to test the hypothesis that the cytochrome P-450 (CYP450) metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) contributes to the afferent arteriolar response to P2 receptor activation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 94-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-81 11668070-1 2001 This study was conducted to test the hypothesis that the cytochrome P-450 (CYP450) metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) contributes to the afferent arteriolar response to P2 receptor activation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 127-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 11668070-1 2001 This study was conducted to test the hypothesis that the cytochrome P-450 (CYP450) metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) contributes to the afferent arteriolar response to P2 receptor activation. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 127-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-81 11668070-9 2001 Taken together, these results are consistent with the hypothesis that the CYP450 metabolite 20-HETE participates in the afferent arteriolar response to activation of P2X receptors. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 92-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-80 11600130-2 2001 The relationship between the structure of N-alkylnitrosamines and CYP form(s) involved in the activation was evaluated. n-alkylnitrosamines 42-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 11725960-3 2001 These oxidations required NADPH, and were markedly inhibited by SKF-525A, indicating that cytochrome P450 (CYP) was involved. NADP 26-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 11725960-3 2001 These oxidations required NADPH, and were markedly inhibited by SKF-525A, indicating that cytochrome P450 (CYP) was involved. NADP 26-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-110 11600130-6 2001 Mutagen-producing capacity of CYP, as indicated by induced revertants/nmol promutagen/pmol CYP, for an N-alkylnitrosamine was determined for all forms of CYP. n-alkylnitrosamine 103-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 11600130-0 2001 Role of human cytochrome P450 (CYP) in the metabolic activation of N-alkylnitrosamines: application of genetically engineered Salmonella typhimurium YG7108 expressing each form of CYP together with human NADPH-cytochrome P450 reductase. n-alkylnitrosamines 67-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 11600130-0 2001 Role of human cytochrome P450 (CYP) in the metabolic activation of N-alkylnitrosamines: application of genetically engineered Salmonella typhimurium YG7108 expressing each form of CYP together with human NADPH-cytochrome P450 reductase. n-alkylnitrosamines 67-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 11600130-0 2001 Role of human cytochrome P450 (CYP) in the metabolic activation of N-alkylnitrosamines: application of genetically engineered Salmonella typhimurium YG7108 expressing each form of CYP together with human NADPH-cytochrome P450 reductase. n-alkylnitrosamines 67-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 11600130-1 2001 The role of human cytochrome P450 (CYP) in the metabolic activation of N-alkylnitrosamines was examined by Ames test using genetically engineered Salmonella typhimurium (S. typhimurium)YG7108 cells expressing each form of human CYP together with human NADPH-cytochrome P450 reductase (OR). n-alkylnitrosamines 71-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 11600130-1 2001 The role of human cytochrome P450 (CYP) in the metabolic activation of N-alkylnitrosamines was examined by Ames test using genetically engineered Salmonella typhimurium (S. typhimurium)YG7108 cells expressing each form of human CYP together with human NADPH-cytochrome P450 reductase (OR). n-alkylnitrosamines 71-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 11600130-1 2001 The role of human cytochrome P450 (CYP) in the metabolic activation of N-alkylnitrosamines was examined by Ames test using genetically engineered Salmonella typhimurium (S. typhimurium)YG7108 cells expressing each form of human CYP together with human NADPH-cytochrome P450 reductase (OR). n-alkylnitrosamines 71-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 228-231 11600130-6 2001 Mutagen-producing capacity of CYP, as indicated by induced revertants/nmol promutagen/pmol CYP, for an N-alkylnitrosamine was determined for all forms of CYP. n-alkylnitrosamine 103-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 11600130-6 2001 Mutagen-producing capacity of CYP, as indicated by induced revertants/nmol promutagen/pmol CYP, for an N-alkylnitrosamine was determined for all forms of CYP. n-alkylnitrosamine 103-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 11525925-1 2001 Metabolic activation of 1-nitropyrene (1-NP) by human cytochrome P450 (P450) family 1 enzymes co-expressed with NADPH-cytochrome P450 reductase (NPR) in Escherichia coli membranes was investigated. 1-nitropyrene 24-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-85 11669629-7 2001 In addition, treatment of CYP4B1, CYP4F3, and CYP4A5/7 with strong base generated a new, chromatographically distinct, polar heme species with a mass of 632.3 amu rather than 616.2 amu. Heme 125-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-40 11509577-2 2001 Cytochrome P450 (P450) 2D6 was first identified as the polymorphic human debrisoquine hydroxylase and subsequently shown to catalyze the oxidation of a variety of drugs containing a basic nitrogen. Nitrogen 188-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 11679404-1 2001 Since the initial reports that renal cytochrome P450 (CYP) enzymes can metabolize arachidonic acid to substances which affect arterial tone, it has become increasingly clear that CYP enzymes expressed within the cardiovascular system play a crucial role in the modulation of vascular homeostasis. Arachidonic Acid 82-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 11679404-1 2001 Since the initial reports that renal cytochrome P450 (CYP) enzymes can metabolize arachidonic acid to substances which affect arterial tone, it has become increasingly clear that CYP enzymes expressed within the cardiovascular system play a crucial role in the modulation of vascular homeostasis. Arachidonic Acid 82-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 11679404-1 2001 Since the initial reports that renal cytochrome P450 (CYP) enzymes can metabolize arachidonic acid to substances which affect arterial tone, it has become increasingly clear that CYP enzymes expressed within the cardiovascular system play a crucial role in the modulation of vascular homeostasis. Arachidonic Acid 82-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-182 11679404-2 2001 There is strong evidence suggesting that the activation of a CYP epoxygenase in endothelial cells is an essential step in nitric oxide and prostacyclin-independent vasodilatation of several vascular beds, particularly in the heart and kidney. Nitric Oxide 122-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 11679404-2 2001 There is strong evidence suggesting that the activation of a CYP epoxygenase in endothelial cells is an essential step in nitric oxide and prostacyclin-independent vasodilatation of several vascular beds, particularly in the heart and kidney. Epoprostenol 139-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 11679404-3 2001 A smooth muscle CYP omega-hydroxylase, on the other hand, generates a vasoconstrictor eicosanoid that is central to the myogenic response. Eicosanoids 86-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 11679404-4 2001 Moreover, CYP epoxygenase and omega-hydroxylase products, as well as CYP-derived reactive oxygen species, are intracellular signal transduction molecules involved in several signaling cascades affecting numerous cellular processes, including vascular cell proliferation and angiogenesis. Reactive Oxygen Species 81-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 11679404-5 2001 This review summarizes the vascular effects of epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid, both of which are CYP-derived metabolites of arachidonic acid, endogenously generated within endothelial and vascular smooth muscle cells. epoxyeicosatrienoic acids 47-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 11679404-5 2001 This review summarizes the vascular effects of epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid, both of which are CYP-derived metabolites of arachidonic acid, endogenously generated within endothelial and vascular smooth muscle cells. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 77-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 11679404-5 2001 This review summarizes the vascular effects of epoxyeicosatrienoic acids and 20-hydroxyeicosatetraenoic acid, both of which are CYP-derived metabolites of arachidonic acid, endogenously generated within endothelial and vascular smooth muscle cells. Arachidonic Acid 155-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 11713692-0 2001 Alternatives to the oxoferryl porphyrin cation radical as the proposed reactive intermediate of cytochrome P450: two-electron oxidized Fe(III) porphyrin derivatives. oxoferryl porphyrin cation radical 20-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-111 11678785-3 2001 Owing to the difficulties associated with in vivo pharmacokinetic studies in a paediatric population, we explored the in vitro metabolism of cisapride by human cytochrome P450. Cisapride 141-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-175 11673755-5 2001 In the absence of any acquired predisposing factor for phenytoin toxicity, genetic mutations in the cytochrome P450 (CYP) enzymes responsible for phenytoin metabolism (CYP2C9 and CYP2C19) were suspected. Phenytoin 55-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 11673755-5 2001 In the absence of any acquired predisposing factor for phenytoin toxicity, genetic mutations in the cytochrome P450 (CYP) enzymes responsible for phenytoin metabolism (CYP2C9 and CYP2C19) were suspected. Phenytoin 146-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 11673755-5 2001 In the absence of any acquired predisposing factor for phenytoin toxicity, genetic mutations in the cytochrome P450 (CYP) enzymes responsible for phenytoin metabolism (CYP2C9 and CYP2C19) were suspected. Phenytoin 146-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-120 11684362-1 2001 N,N-dimethylformamide (DMF), an organic solvent widely used in industry, is bioactivated by cytochrome P450 (P450) to reactive metabolites which are believed to be responsible for the hepatotoxicity observed in animals and humans. Dimethylformamide 0-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 11684362-1 2001 N,N-dimethylformamide (DMF), an organic solvent widely used in industry, is bioactivated by cytochrome P450 (P450) to reactive metabolites which are believed to be responsible for the hepatotoxicity observed in animals and humans. Dimethylformamide 23-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 11560868-1 2001 The antipsychotic agent risperidone, is metabolized by different cytochrome P-450 (CYP) enzymes, including CYP2D6, to the active 9-hydroxyrisperidone, which is the major metabolite in plasma. Risperidone 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 11560868-1 2001 The antipsychotic agent risperidone, is metabolized by different cytochrome P-450 (CYP) enzymes, including CYP2D6, to the active 9-hydroxyrisperidone, which is the major metabolite in plasma. Risperidone 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 11560868-1 2001 The antipsychotic agent risperidone, is metabolized by different cytochrome P-450 (CYP) enzymes, including CYP2D6, to the active 9-hydroxyrisperidone, which is the major metabolite in plasma. Paliperidone Palmitate 129-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 11560868-1 2001 The antipsychotic agent risperidone, is metabolized by different cytochrome P-450 (CYP) enzymes, including CYP2D6, to the active 9-hydroxyrisperidone, which is the major metabolite in plasma. Paliperidone Palmitate 129-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 11713692-0 2001 Alternatives to the oxoferryl porphyrin cation radical as the proposed reactive intermediate of cytochrome P450: two-electron oxidized Fe(III) porphyrin derivatives. fe(iii) porphyrin 135-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-111 11551516-0 2001 Relationships between non-occupational cadmium exposure and expression of nine cytochrome P450 forms in human liver and kidney cortex samples. Cadmium 39-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 11601668-3 2001 Like nonselective NSAIDs, coxibs are hepatically metabolized: rofecoxib primarily by reduction by cytosolic enzymes and celecoxib by the cytochrome P450 (CYP) enzyme system. rofecoxib 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-152 11601668-3 2001 Like nonselective NSAIDs, coxibs are hepatically metabolized: rofecoxib primarily by reduction by cytosolic enzymes and celecoxib by the cytochrome P450 (CYP) enzyme system. rofecoxib 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-157 11601668-3 2001 Like nonselective NSAIDs, coxibs are hepatically metabolized: rofecoxib primarily by reduction by cytosolic enzymes and celecoxib by the cytochrome P450 (CYP) enzyme system. Celecoxib 120-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-152 11601668-3 2001 Like nonselective NSAIDs, coxibs are hepatically metabolized: rofecoxib primarily by reduction by cytosolic enzymes and celecoxib by the cytochrome P450 (CYP) enzyme system. Celecoxib 120-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-157 11601668-5 2001 However, potent inducers of CYP, such as rifampin, may decrease rofecoxib concentrations because of induction of general hepatic metabolic activity. Rifampin 41-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 11601668-5 2001 However, potent inducers of CYP, such as rifampin, may decrease rofecoxib concentrations because of induction of general hepatic metabolic activity. rofecoxib 64-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 11586481-3 2001 Modulation of xenobiotic metabolism, including cytochrome P-450 (CYP) enzyme activities, due to dietary intakes of cruciferous vegetables, has been described in animals and humans, and the induction of CYP1A enzymes is suggested mainly to be related to the content of indolyl glucosinolates in these vegetables. indolyl glucosinolates 268-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 11516890-3 2001 Although metabolized by the cytochrome P-450 (CYP) system, venlafaxine inhibits CYP 2D6 and 3A4 to a far lesser extent than do the SSRIs. Venlafaxine Hydrochloride 59-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 11551516-1 2001 This study was undertaken to assess associations between age, gender, cigarette smoke and non-workplace cadmium exposure, and liver pathology and inter-individual variation in cytochrome P450 (CYP) expression in human tissues. Cadmium 104-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-196 11502591-0 2001 Role of guanylyl cyclase and cytochrome P-450 on renal response to nitric oxide. Nitric Oxide 67-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 11556812-1 2001 Cytochrome P450 (P450) 2D6 is a polymorphic human enzyme involved in the oxidation of >50 drugs, most of which contain a basic nitrogen. Nitrogen 130-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 11470492-0 2001 Inhibition by green tea catechins of metabolic activation of procarcinogens by human cytochrome P450. Catechin 24-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 11678058-5 2001 A review of recent literature suggests that clarithromycin may induce digoxin toxicity by three different mechanisms, including reduction of renal excretion of digoxin, alteration of intestinal flora, and inhibition of cytochrome P-450 in the liver. Clarithromycin 44-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 219-235 11502728-0 2001 Metabolism of chlorpyrifos by human cytochrome P450 isoforms and human, mouse, and rat liver microsomes. Chlorpyrifos 14-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 11502728-2 2001 CPS can be activated by cytochrome P450 (CYP) through a desulfuration reaction to form chlorpyrifos-oxon (CPO), a potent anticholinesterase. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 87-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 11502728-2 2001 CPS can be activated by cytochrome P450 (CYP) through a desulfuration reaction to form chlorpyrifos-oxon (CPO), a potent anticholinesterase. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 87-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 11502728-2 2001 CPS can be activated by cytochrome P450 (CYP) through a desulfuration reaction to form chlorpyrifos-oxon (CPO), a potent anticholinesterase. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 106-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 11502728-2 2001 CPS can be activated by cytochrome P450 (CYP) through a desulfuration reaction to form chlorpyrifos-oxon (CPO), a potent anticholinesterase. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 106-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-44 11497333-9 2001 Cells pretreated with 50 degrees M 3-methylcholanthrene (3MC), a CYP 1A inducer, for 72 h prior to 0.15 microM AFB1, produced the activated AFB1 8,9-epoxide (AFBO). Methylcholanthrene 35-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 11497333-9 2001 Cells pretreated with 50 degrees M 3-methylcholanthrene (3MC), a CYP 1A inducer, for 72 h prior to 0.15 microM AFB1, produced the activated AFB1 8,9-epoxide (AFBO). Methylcholanthrene 57-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 11497333-9 2001 Cells pretreated with 50 degrees M 3-methylcholanthrene (3MC), a CYP 1A inducer, for 72 h prior to 0.15 microM AFB1, produced the activated AFB1 8,9-epoxide (AFBO). afb1 8,9-epoxide 140-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 11497333-13 2001 Expression of CYP 1A was significantly increased in 3MC-pretreated cells, while CYP 3A4 expression increased slightly, but not to the extent of the 1A isoforms. Methylcholanthrene 52-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 11497333-17 2001 These data support earlier findings showing modest CYP-mediated AFB1 activation in human airways, but indicate that exposure to polycyclic aromatic hydrocarbons (PAHs), such as 3MC, which induce CYP(s) that specifically activate AFB1 may increase the harmful effects of AFB1 exposures in human airways. Polycyclic Aromatic Hydrocarbons 128-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-198 11497333-17 2001 These data support earlier findings showing modest CYP-mediated AFB1 activation in human airways, but indicate that exposure to polycyclic aromatic hydrocarbons (PAHs), such as 3MC, which induce CYP(s) that specifically activate AFB1 may increase the harmful effects of AFB1 exposures in human airways. Polycyclic Aromatic Hydrocarbons 162-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-198 11497333-17 2001 These data support earlier findings showing modest CYP-mediated AFB1 activation in human airways, but indicate that exposure to polycyclic aromatic hydrocarbons (PAHs), such as 3MC, which induce CYP(s) that specifically activate AFB1 may increase the harmful effects of AFB1 exposures in human airways. Methylcholanthrene 177-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-198 11502723-0 2001 In vitro characterization of the inhibition profile of loratadine, desloratadine, and 3-OH-desloratadine for five human cytochrome P-450 enzymes. Loratadine 55-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 11502723-0 2001 In vitro characterization of the inhibition profile of loratadine, desloratadine, and 3-OH-desloratadine for five human cytochrome P-450 enzymes. desloratadine 67-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 11502723-0 2001 In vitro characterization of the inhibition profile of loratadine, desloratadine, and 3-OH-desloratadine for five human cytochrome P-450 enzymes. 3-oh-desloratadine 86-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 11502723-1 2001 The purpose of this study was to evaluate loratadine, desloratadine, and 3-OH-desloratadine as inhibitors of certain human liver cytochrome P-450 enzymes. Loratadine 42-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 11502723-1 2001 The purpose of this study was to evaluate loratadine, desloratadine, and 3-OH-desloratadine as inhibitors of certain human liver cytochrome P-450 enzymes. desloratadine 54-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 11502723-1 2001 The purpose of this study was to evaluate loratadine, desloratadine, and 3-OH-desloratadine as inhibitors of certain human liver cytochrome P-450 enzymes. 3-oh-desloratadine 73-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 11506821-6 2001 Earlier in vivo experiments with rodents indicated that indoles and isothiocyanates, two major groups of glucosinolate breakdown products, attenuate the effects of polycyclic aromatic hydrocarbons (PAHs) and nitrosamines via induction of GST and inhibition of cytochrome-P450 isoenzymes, respectively. Indoles 56-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 260-275 11506821-6 2001 Earlier in vivo experiments with rodents indicated that indoles and isothiocyanates, two major groups of glucosinolate breakdown products, attenuate the effects of polycyclic aromatic hydrocarbons (PAHs) and nitrosamines via induction of GST and inhibition of cytochrome-P450 isoenzymes, respectively. Isothiocyanates 68-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 260-275 11506821-6 2001 Earlier in vivo experiments with rodents indicated that indoles and isothiocyanates, two major groups of glucosinolate breakdown products, attenuate the effects of polycyclic aromatic hydrocarbons (PAHs) and nitrosamines via induction of GST and inhibition of cytochrome-P450 isoenzymes, respectively. Polycyclic Aromatic Hydrocarbons 164-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 260-275 11470492-6 2001 The IC(50) values of catechins for the inhibition of human CYP were roughly the same as those seen in the mutagenic activation. Catechin 21-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 11470492-7 2001 EGCG inhibited other forms of human CYP such as CYP2A6, CYP2C19 and CYP2E1, indicating the non-specific inhibitory effects of EGCG toward human CYPs. epigallocatechin gallate 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 11470492-7 2001 EGCG inhibited other forms of human CYP such as CYP2A6, CYP2C19 and CYP2E1, indicating the non-specific inhibitory effects of EGCG toward human CYPs. epigallocatechin gallate 126-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 11470492-8 2001 Furthermore, EGCG inhibited human NADPH-cytochrome CYP reductase (OR) with a K(i) value of 2.5 microM. epigallocatechin gallate 13-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 11454723-1 2001 We examined the effects of several agents, including dietary flavonoids, on CYP1A1 expression utilizing a recently developed high-throughput screening system for assessing human cytochrome P450 (CYP) induction. Flavonoids 61-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 11500893-0 2001 How Does Ethene Inactivate Cytochrome P450 En Route to Its Epoxidation? ethylene 9-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 29711977-0 2001 How Does Ethene Inactivate Cytochrome P450 En Route to Its Epoxidation? ethylene 9-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 29711977-2 2001 The suicidal complex 4 2, which inactivates cytochrome P450 during olefin epoxidation, was shown by density functional calculations to be formed from the same high-spin intermediate (4 1-III) that leads to stereochemical scrambling. Alkenes 67-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 11504269-0 2001 Contributions of five human cytochrome P450 isoforms to the N-demethylation of clozapine in vitro at low and high concentrations. Clozapine 79-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 11454729-0 2001 Inhibition and inactivation of human cytochrome P450 isoforms by phenethyl isothiocyanate. phenethyl isothiocyanate 65-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 11454729-1 2001 The inhibition and mechanism-based inactivation potencies of phenethyl isothiocyanate (PEITC) for human cytochrome P450 (CYP) activities were investigated using microsomes from baculovirus-infected insect cells expressing specific human CYP isoforms. phenethyl isothiocyanate 61-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 11454729-1 2001 The inhibition and mechanism-based inactivation potencies of phenethyl isothiocyanate (PEITC) for human cytochrome P450 (CYP) activities were investigated using microsomes from baculovirus-infected insect cells expressing specific human CYP isoforms. phenethyl isothiocyanate 61-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 11454729-1 2001 The inhibition and mechanism-based inactivation potencies of phenethyl isothiocyanate (PEITC) for human cytochrome P450 (CYP) activities were investigated using microsomes from baculovirus-infected insect cells expressing specific human CYP isoforms. phenethyl isothiocyanate 61-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 237-240 11454729-1 2001 The inhibition and mechanism-based inactivation potencies of phenethyl isothiocyanate (PEITC) for human cytochrome P450 (CYP) activities were investigated using microsomes from baculovirus-infected insect cells expressing specific human CYP isoforms. phenethyl isothiocyanate 87-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 11454729-1 2001 The inhibition and mechanism-based inactivation potencies of phenethyl isothiocyanate (PEITC) for human cytochrome P450 (CYP) activities were investigated using microsomes from baculovirus-infected insect cells expressing specific human CYP isoforms. phenethyl isothiocyanate 87-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 11454729-1 2001 The inhibition and mechanism-based inactivation potencies of phenethyl isothiocyanate (PEITC) for human cytochrome P450 (CYP) activities were investigated using microsomes from baculovirus-infected insect cells expressing specific human CYP isoforms. phenethyl isothiocyanate 87-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 237-240 11454729-10 2001 The present study directly proved that the chemopreventive effects of PEITC for nitrosamine-induced carcinogenesis are due to the inhibition of CYP by an in vitro study. phenethyl isothiocyanate 70-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-147 11454729-10 2001 The present study directly proved that the chemopreventive effects of PEITC for nitrosamine-induced carcinogenesis are due to the inhibition of CYP by an in vitro study. Nitrosamines 80-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-147 11454729-11 2001 The possibility that PEITC would affect the pharmacokinetics of clinically used drugs that are metabolized by these CYP isoforms was also suggested. phenethyl isothiocyanate 21-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-119 11454734-0 2001 Identification of cytochrome P-450 isoforms responsible for cis-tramadol metabolism in human liver microsomes. Tramadol 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 11454734-1 2001 The metabolism of cis-tramadol has been studied in human liver microsomes and in cDNA-expressed human cytochrome P-450 (CYP) isoforms. Tramadol 18-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 11454734-1 2001 The metabolism of cis-tramadol has been studied in human liver microsomes and in cDNA-expressed human cytochrome P-450 (CYP) isoforms. Tramadol 18-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 11454734-4 2001 Kinetic analysis revealed that multiple CYP enzymes were involved in the metabolism of tramadol to both M1 and M2. Tramadol 87-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 11454734-11 2001 By kinetic analysis and the results of the reaction phenotyping studies, tramadol metabolism in human liver is catalyzed by multiple CYP isoforms. Tramadol 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 11504269-1 2001 The authors assessed the in vitro contribution of cytochrome P450 (CYP) isoforms 1A2, 3A4, 2C9, 2C19, and 2D6 to the N-demethylation of clozapine mediated by human liver microsomal preparations (HLM). Nitrogen 117-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 11504269-1 2001 The authors assessed the in vitro contribution of cytochrome P450 (CYP) isoforms 1A2, 3A4, 2C9, 2C19, and 2D6 to the N-demethylation of clozapine mediated by human liver microsomal preparations (HLM). Nitrogen 117-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11504269-1 2001 The authors assessed the in vitro contribution of cytochrome P450 (CYP) isoforms 1A2, 3A4, 2C9, 2C19, and 2D6 to the N-demethylation of clozapine mediated by human liver microsomal preparations (HLM). Clozapine 136-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 11504269-1 2001 The authors assessed the in vitro contribution of cytochrome P450 (CYP) isoforms 1A2, 3A4, 2C9, 2C19, and 2D6 to the N-demethylation of clozapine mediated by human liver microsomal preparations (HLM). Clozapine 136-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11452704-0 2001 Lack of gender differences and large intrasubject variability in cytochrome P450 activity measured by phenotyping with dextromethorphan. Dextromethorphan 119-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 11427068-2 2001 An initial model compound was constructed from a structure obtained by 300-ps molecular dynamics simulation of compound I-formed P-450cam under physiologic conditions, and it consisted of porphine for protoporphyrin IX, S(-)-CH(3) for the side chain of Cys357 of the fifth ligand of heme, a methane molecule for the substrate, a heme iron, and an oxygen atom of the sixth ligand of heme. porphine 188-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-121 11427068-2 2001 An initial model compound was constructed from a structure obtained by 300-ps molecular dynamics simulation of compound I-formed P-450cam under physiologic conditions, and it consisted of porphine for protoporphyrin IX, S(-)-CH(3) for the side chain of Cys357 of the fifth ligand of heme, a methane molecule for the substrate, a heme iron, and an oxygen atom of the sixth ligand of heme. Heme 283-287 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-121 11427068-2 2001 An initial model compound was constructed from a structure obtained by 300-ps molecular dynamics simulation of compound I-formed P-450cam under physiologic conditions, and it consisted of porphine for protoporphyrin IX, S(-)-CH(3) for the side chain of Cys357 of the fifth ligand of heme, a methane molecule for the substrate, a heme iron, and an oxygen atom of the sixth ligand of heme. Methane 291-298 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-121 11427068-2 2001 An initial model compound was constructed from a structure obtained by 300-ps molecular dynamics simulation of compound I-formed P-450cam under physiologic conditions, and it consisted of porphine for protoporphyrin IX, S(-)-CH(3) for the side chain of Cys357 of the fifth ligand of heme, a methane molecule for the substrate, a heme iron, and an oxygen atom of the sixth ligand of heme. Heme 329-333 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-121 11427068-2 2001 An initial model compound was constructed from a structure obtained by 300-ps molecular dynamics simulation of compound I-formed P-450cam under physiologic conditions, and it consisted of porphine for protoporphyrin IX, S(-)-CH(3) for the side chain of Cys357 of the fifth ligand of heme, a methane molecule for the substrate, a heme iron, and an oxygen atom of the sixth ligand of heme. Iron 334-338 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-121 11427068-2 2001 An initial model compound was constructed from a structure obtained by 300-ps molecular dynamics simulation of compound I-formed P-450cam under physiologic conditions, and it consisted of porphine for protoporphyrin IX, S(-)-CH(3) for the side chain of Cys357 of the fifth ligand of heme, a methane molecule for the substrate, a heme iron, and an oxygen atom of the sixth ligand of heme. Oxygen 347-353 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-121 11427068-2 2001 An initial model compound was constructed from a structure obtained by 300-ps molecular dynamics simulation of compound I-formed P-450cam under physiologic conditions, and it consisted of porphine for protoporphyrin IX, S(-)-CH(3) for the side chain of Cys357 of the fifth ligand of heme, a methane molecule for the substrate, a heme iron, and an oxygen atom of the sixth ligand of heme. Heme 329-333 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-121 11488523-1 2001 Several recent studies have demonstrated that the cytochrome p450 (CYP) family plays an important role in the metabolism of taxanes. Taxoids 124-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 11488523-1 2001 Several recent studies have demonstrated that the cytochrome p450 (CYP) family plays an important role in the metabolism of taxanes. Taxoids 124-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11408374-0 2001 Cytochrome P450-catalyzed metabolism of ezlopitant alkene (CJ-12,458), a pharmacologically active metabolite of ezlopitant: enzyme kinetics and mechanism of an alkene hydration reaction. Alkenes 51-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11408374-0 2001 Cytochrome P450-catalyzed metabolism of ezlopitant alkene (CJ-12,458), a pharmacologically active metabolite of ezlopitant: enzyme kinetics and mechanism of an alkene hydration reaction. cj-12 59-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11408374-0 2001 Cytochrome P450-catalyzed metabolism of ezlopitant alkene (CJ-12,458), a pharmacologically active metabolite of ezlopitant: enzyme kinetics and mechanism of an alkene hydration reaction. Alkenes 160-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11408374-2 2001 In incubations with human liver microsomes and cofactors required for cytochrome P450 (CYP) activity, CJ-12,458 was converted to two metabolites: a diol (CP-611,781) and a 1 degrees alcohol (CP-616,762). cj-12 102-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 11408374-2 2001 In incubations with human liver microsomes and cofactors required for cytochrome P450 (CYP) activity, CJ-12,458 was converted to two metabolites: a diol (CP-611,781) and a 1 degrees alcohol (CP-616,762). cj-12 102-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 11408374-2 2001 In incubations with human liver microsomes and cofactors required for cytochrome P450 (CYP) activity, CJ-12,458 was converted to two metabolites: a diol (CP-611,781) and a 1 degrees alcohol (CP-616,762). diol 148-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 11408374-2 2001 In incubations with human liver microsomes and cofactors required for cytochrome P450 (CYP) activity, CJ-12,458 was converted to two metabolites: a diol (CP-611,781) and a 1 degrees alcohol (CP-616,762). diol 148-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 11408374-2 2001 In incubations with human liver microsomes and cofactors required for cytochrome P450 (CYP) activity, CJ-12,458 was converted to two metabolites: a diol (CP-611,781) and a 1 degrees alcohol (CP-616,762). Alcohols 182-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 11408374-2 2001 In incubations with human liver microsomes and cofactors required for cytochrome P450 (CYP) activity, CJ-12,458 was converted to two metabolites: a diol (CP-611,781) and a 1 degrees alcohol (CP-616,762). Alcohols 182-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 11356600-10 2001 In the human forearm microvasculature, EDHF appears to be a cytochrome P-450 derivative, possibly an epoxyeicosatrienoic acid. edhf 39-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 11343389-0 2001 Extreme warfarin sensitivity in siblings associated with multiple cytochrome P450 polymorphisms. Warfarin 8-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 11343389-2 2001 Warfarin is metabolized by two distinct subfamilies of the cytochrome P450 (CYP) complex. Warfarin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 11343389-2 2001 Warfarin is metabolized by two distinct subfamilies of the cytochrome P450 (CYP) complex. Warfarin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 11343389-3 2001 We describe two siblings with extreme sensitivity to warfarin who share an unusual CYP genotype. Warfarin 53-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 11356600-10 2001 In the human forearm microvasculature, EDHF appears to be a cytochrome P-450 derivative, possibly an epoxyeicosatrienoic acid. epoxyeicosatrienoic acid 101-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 11353747-6 2001 Furthermore, P450 MI-2 and P450 MI-3 were recognized by anti-CYP4F and anti-CYP3A antibodies, respectively, in immunoblot analysis and catalyzed leukotriene B(4) omega-hydroxylation and testosterone 6beta-hydroxylation, which are known to be mediated by CYP4F and CYP3A, respectively. Leukotriene B4 145-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-66 11375247-1 2001 The present study examined the inhibitory effects of N-hydroxy-N"-(4-butyl-2-methylphenyl)-formamidine (HET0016) on the renal metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes. n-hydroxy-n"-(4-butyl-2-methylphenyl)-formamidine 53-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-175 11375247-1 2001 The present study examined the inhibitory effects of N-hydroxy-N"-(4-butyl-2-methylphenyl)-formamidine (HET0016) on the renal metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes. n-hydroxy-n"-(4-butyl-2-methylphenyl)-formamidine 53-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 11375247-1 2001 The present study examined the inhibitory effects of N-hydroxy-N"-(4-butyl-2-methylphenyl)-formamidine (HET0016) on the renal metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes. Arachidonic Acid 140-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-175 11375247-1 2001 The present study examined the inhibitory effects of N-hydroxy-N"-(4-butyl-2-methylphenyl)-formamidine (HET0016) on the renal metabolism of arachidonic acid by cytochrome P450 (CYP) enzymes. Arachidonic Acid 140-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 11497349-6 2001 The use of antisense morpholino oligonucleotide strategies to target CYP enzymes may result in safer and more uniform therapeutic applications. Morpholinos 21-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 11353758-1 2001 Ketamine is metabolized by cytochrome P450 (CYP) leading to production of pharmacologically active products and contributing to drug excretion. Ketamine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 11353758-1 2001 Ketamine is metabolized by cytochrome P450 (CYP) leading to production of pharmacologically active products and contributing to drug excretion. Ketamine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 11353758-2 2001 We identified the CYP enzymes involved in the N-demethylation of ketamine enantiomers using pooled human liver microsomes and microsomes from human B-lymphoblastoid cells that expressed CYP enzymes. Nitrogen 46-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 11353758-2 2001 We identified the CYP enzymes involved in the N-demethylation of ketamine enantiomers using pooled human liver microsomes and microsomes from human B-lymphoblastoid cells that expressed CYP enzymes. Nitrogen 46-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-189 11498765-1 2001 The sensitivity of tumors to cyclophosphamide (CPA) and other anticancer prodrugs can be substantially enhanced by transduction of tumors with a prodrug-activating mammalian cytochrome P450 (CYP) enzyme in combination with the flavoenzyme P450 reductase. Cyclophosphamide 29-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-189 11498765-1 2001 The sensitivity of tumors to cyclophosphamide (CPA) and other anticancer prodrugs can be substantially enhanced by transduction of tumors with a prodrug-activating mammalian cytochrome P450 (CYP) enzyme in combination with the flavoenzyme P450 reductase. Cyclophosphamide 29-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-194 11498765-1 2001 The sensitivity of tumors to cyclophosphamide (CPA) and other anticancer prodrugs can be substantially enhanced by transduction of tumors with a prodrug-activating mammalian cytochrome P450 (CYP) enzyme in combination with the flavoenzyme P450 reductase. Cyclophosphamide 47-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-189 11498765-1 2001 The sensitivity of tumors to cyclophosphamide (CPA) and other anticancer prodrugs can be substantially enhanced by transduction of tumors with a prodrug-activating mammalian cytochrome P450 (CYP) enzyme in combination with the flavoenzyme P450 reductase. Cyclophosphamide 47-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-194 11353747-6 2001 Furthermore, P450 MI-2 and P450 MI-3 were recognized by anti-CYP4F and anti-CYP3A antibodies, respectively, in immunoblot analysis and catalyzed leukotriene B(4) omega-hydroxylation and testosterone 6beta-hydroxylation, which are known to be mediated by CYP4F and CYP3A, respectively. testosterone 6beta 186-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-66 11353747-9 2001 These results indicate that P450 MI-2 belongs to the CYP4F subfamily and is mainly responsible for hydroxylation of ebastine in monkey small intestinal microsomes. ebastine 116-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-58 11353749-8 2001 Cytochrome P450 (P450) reaction phenotyping studies-using P450 form selective chemical inhibitors, immunoinhibitory antibodies, recombinant P450s, and correlation analysis with microsomes prepared from a bank of human livers-revealed that the 6"-methyl hydroxylation of etoricoxib was catalyzed largely (approximately 60%) by member(s) of the CYP3A subfamily. Etoricoxib 270-280 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11353758-2 2001 We identified the CYP enzymes involved in the N-demethylation of ketamine enantiomers using pooled human liver microsomes and microsomes from human B-lymphoblastoid cells that expressed CYP enzymes. Ketamine 65-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 11353758-2 2001 We identified the CYP enzymes involved in the N-demethylation of ketamine enantiomers using pooled human liver microsomes and microsomes from human B-lymphoblastoid cells that expressed CYP enzymes. Ketamine 65-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-189 11353747-6 2001 Furthermore, P450 MI-2 and P450 MI-3 were recognized by anti-CYP4F and anti-CYP3A antibodies, respectively, in immunoblot analysis and catalyzed leukotriene B(4) omega-hydroxylation and testosterone 6beta-hydroxylation, which are known to be mediated by CYP4F and CYP3A, respectively. Leukotriene B4 145-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 254-259 11353758-5 2001 Among the 12 cDNA-expressed CYP enzymes examined, CYP2B6, CYP2C9, and CYP3A4 showed high activities for the N-demethylation of both enantiomers at the substrate concentration of 1 mM. Nitrogen 15-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 11468938-0 2001 A mechanism-based pharmacokinetic model for the cytochrome P450 drug-drug interaction between cyclophosphamide and thioTEPA and the autoinduction of cyclophosphamide. Cyclophosphamide 94-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 11468938-0 2001 A mechanism-based pharmacokinetic model for the cytochrome P450 drug-drug interaction between cyclophosphamide and thioTEPA and the autoinduction of cyclophosphamide. Thiotepa 115-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 11468938-0 2001 A mechanism-based pharmacokinetic model for the cytochrome P450 drug-drug interaction between cyclophosphamide and thioTEPA and the autoinduction of cyclophosphamide. Cyclophosphamide 149-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 11468938-2 2001 CP is a prodrug and is activated by cytochrome P450 to 4-hydroxycyclophosphamide (HCP) which yields the final cytotoxic metabolite phosphoramide mustard (PM). 4-hydroxycyclophosphamide 55-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 11468938-2 2001 CP is a prodrug and is activated by cytochrome P450 to 4-hydroxycyclophosphamide (HCP) which yields the final cytotoxic metabolite phosphoramide mustard (PM). 4-hydroxycyclophosphamide 82-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 11468938-2 2001 CP is a prodrug and is activated by cytochrome P450 to 4-hydroxycyclophosphamide (HCP) which yields the final cytotoxic metabolite phosphoramide mustard (PM). phosphoramide mustard 131-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 11377097-9 2001 These results show the metabolic activation of APAP, CPA and BA by the specific CYP subtypes expressed in the transformants and demonstrate the usefulness of these transformants for in vitro metabolic and toxicological studies in human liver. Cyclophosphamide 53-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 11377097-9 2001 These results show the metabolic activation of APAP, CPA and BA by the specific CYP subtypes expressed in the transformants and demonstrate the usefulness of these transformants for in vitro metabolic and toxicological studies in human liver. benz(a)anthracene 61-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 11377097-9 2001 These results show the metabolic activation of APAP, CPA and BA by the specific CYP subtypes expressed in the transformants and demonstrate the usefulness of these transformants for in vitro metabolic and toxicological studies in human liver. Acetaminophen 47-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 11712183-0 2001 Effect of adamantanone derivative possessing anti-HIV properties on the redox processes in the cytochrome P-450 system. adamantanone 10-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 11336973-3 2001 Aroclor 1254-induced and uninduced rat liver microsomes were compared to human liver microsomes in the metabolism of substrates which are known to be selectively metabolized by the major human cytochrome P450 (CYP) isoforms. Chlorodiphenyl (54% Chlorine) 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 193-208 11336973-3 2001 Aroclor 1254-induced and uninduced rat liver microsomes were compared to human liver microsomes in the metabolism of substrates which are known to be selectively metabolized by the major human cytochrome P450 (CYP) isoforms. Chlorodiphenyl (54% Chlorine) 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 210-213 11430508-4 2001 Rabeprazole and esomeprazole achieve more rapid and profound inhibition of acid secretion than do older agents, and they sustain this suppression to provide acid control and symptom relief over 24 h. The balanced hepatic metabolism of rabeprazole, involving both cytochrome P450 (CYP)-mediated reactions in the liver and nonenzymatic reactions, appears to confer an advantage over older PPIs in that genetic polymorphisms for CYP 2C19 do not significantly influence rabeprazole clearance and, potentially, clinical efficacy. Rabeprazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 263-278 11430508-4 2001 Rabeprazole and esomeprazole achieve more rapid and profound inhibition of acid secretion than do older agents, and they sustain this suppression to provide acid control and symptom relief over 24 h. The balanced hepatic metabolism of rabeprazole, involving both cytochrome P450 (CYP)-mediated reactions in the liver and nonenzymatic reactions, appears to confer an advantage over older PPIs in that genetic polymorphisms for CYP 2C19 do not significantly influence rabeprazole clearance and, potentially, clinical efficacy. Rabeprazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 280-283 11430508-4 2001 Rabeprazole and esomeprazole achieve more rapid and profound inhibition of acid secretion than do older agents, and they sustain this suppression to provide acid control and symptom relief over 24 h. The balanced hepatic metabolism of rabeprazole, involving both cytochrome P450 (CYP)-mediated reactions in the liver and nonenzymatic reactions, appears to confer an advantage over older PPIs in that genetic polymorphisms for CYP 2C19 do not significantly influence rabeprazole clearance and, potentially, clinical efficacy. Esomeprazole 16-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 263-278 11430508-4 2001 Rabeprazole and esomeprazole achieve more rapid and profound inhibition of acid secretion than do older agents, and they sustain this suppression to provide acid control and symptom relief over 24 h. The balanced hepatic metabolism of rabeprazole, involving both cytochrome P450 (CYP)-mediated reactions in the liver and nonenzymatic reactions, appears to confer an advantage over older PPIs in that genetic polymorphisms for CYP 2C19 do not significantly influence rabeprazole clearance and, potentially, clinical efficacy. Esomeprazole 16-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 280-283 11430508-4 2001 Rabeprazole and esomeprazole achieve more rapid and profound inhibition of acid secretion than do older agents, and they sustain this suppression to provide acid control and symptom relief over 24 h. The balanced hepatic metabolism of rabeprazole, involving both cytochrome P450 (CYP)-mediated reactions in the liver and nonenzymatic reactions, appears to confer an advantage over older PPIs in that genetic polymorphisms for CYP 2C19 do not significantly influence rabeprazole clearance and, potentially, clinical efficacy. Rabeprazole 235-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 263-278 11430508-4 2001 Rabeprazole and esomeprazole achieve more rapid and profound inhibition of acid secretion than do older agents, and they sustain this suppression to provide acid control and symptom relief over 24 h. The balanced hepatic metabolism of rabeprazole, involving both cytochrome P450 (CYP)-mediated reactions in the liver and nonenzymatic reactions, appears to confer an advantage over older PPIs in that genetic polymorphisms for CYP 2C19 do not significantly influence rabeprazole clearance and, potentially, clinical efficacy. Rabeprazole 235-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 280-283 11476153-1 2001 The aim of the present study was to assess the interindividual variation in styrene toxicokinetics and to correlate this variation with the individual metabolic capacity for cytochrome P450 (CYP), CYP2E1, CYP1A2 and CYP2D6. Styrene 76-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-189 11476153-1 2001 The aim of the present study was to assess the interindividual variation in styrene toxicokinetics and to correlate this variation with the individual metabolic capacity for cytochrome P450 (CYP), CYP2E1, CYP1A2 and CYP2D6. Styrene 76-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 191-194 11259320-0 2001 Metabolism of dimethyl-4,4"-dimethoxy-5,6,5",6"-dimethylene dioxybiphenyl-2,2"-dicarboxylate (DDB) by human liver microsomes: characterization of metabolic pathways and of cytochrome P450 isoforms involved. dimethyl-4,4'-dimethoxy-5,6,5',6'-dimethylene dioxybiphenyl-2,2'-dicarboxylate 14-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-187 11504148-0 2001 Human cytochrome P450 isozymes in metabolism and health effects of gasoline ethers. gasoline ethers 67-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 11504148-7 2001 Formation of TBA in human liver microsomes is NADPH-dependent and is significantly inhibited by carbon monoxide, which inhibits cytochrome P450 (CYP) enzymes. tert-Butyl Alcohol 13-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-143 11504148-7 2001 Formation of TBA in human liver microsomes is NADPH-dependent and is significantly inhibited by carbon monoxide, which inhibits cytochrome P450 (CYP) enzymes. tert-Butyl Alcohol 13-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 11504148-7 2001 Formation of TBA in human liver microsomes is NADPH-dependent and is significantly inhibited by carbon monoxide, which inhibits cytochrome P450 (CYP) enzymes. NADP 46-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 11504148-7 2001 Formation of TBA in human liver microsomes is NADPH-dependent and is significantly inhibited by carbon monoxide, which inhibits cytochrome P450 (CYP) enzymes. Carbon Monoxide 96-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-143 11504148-7 2001 Formation of TBA in human liver microsomes is NADPH-dependent and is significantly inhibited by carbon monoxide, which inhibits cytochrome P450 (CYP) enzymes. Carbon Monoxide 96-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 11504148-13 2001 Correlation analysis of the ether-metabolizing activities with individual CYP enzyme activities in the human liver microsomes showed that the highest degree of correlation was with CYP isoform 2A6 (CYP2A6)+ (r = 0.94 for MTBE, 0.95 for ETBE, and 0.90 for TAME), which is constitutively expressed in human livers and known to be polymorphic. Ether 28-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 11504148-14 2001 CYP2A6 displayed the highest turnover number in metabolizing gasoline ethers among a battery of human CYP enzymes expressed in human B-lymphoblastoid cells. Ethers 70-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 11311457-1 2001 The bioactivation of N-nitrosamines and polycyclic aromatic hydrocarbons (PAHs) is mediated primarily by the mixed-function oxidase system, which includes dimethylnitrosamine N-demethylase I, arylhydrocarbon [benzo(a)pyerne] hydroxylase, cytochrome P450, cytochrome b(5), and ethoxycoumarin deethylase. n-nitrosamines 21-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 238-269 11311457-1 2001 The bioactivation of N-nitrosamines and polycyclic aromatic hydrocarbons (PAHs) is mediated primarily by the mixed-function oxidase system, which includes dimethylnitrosamine N-demethylase I, arylhydrocarbon [benzo(a)pyerne] hydroxylase, cytochrome P450, cytochrome b(5), and ethoxycoumarin deethylase. Polycyclic Aromatic Hydrocarbons 40-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 238-269 11311457-1 2001 The bioactivation of N-nitrosamines and polycyclic aromatic hydrocarbons (PAHs) is mediated primarily by the mixed-function oxidase system, which includes dimethylnitrosamine N-demethylase I, arylhydrocarbon [benzo(a)pyerne] hydroxylase, cytochrome P450, cytochrome b(5), and ethoxycoumarin deethylase. Polycyclic Aromatic Hydrocarbons 74-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 238-269 11457014-0 2001 Multi-state epoxidation of ethene by cytochrome P450: a quantum chemical study. ethylene 27-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 11457014-1 2001 The epoxidation of ethene by a model for Compound I of cytochrome P450, studied by the use of density functional B3LYP calculations, involves two-state reactivity (TSR) with multiple electromer species, hence "multi-state epoxidation". ethylene 19-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 11259320-0 2001 Metabolism of dimethyl-4,4"-dimethoxy-5,6,5",6"-dimethylene dioxybiphenyl-2,2"-dicarboxylate (DDB) by human liver microsomes: characterization of metabolic pathways and of cytochrome P450 isoforms involved. ddb 94-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-187 11259320-7 2001 Cytochrome P450 (P450) isoform(s) involved in the metabolism of DDB was identified using several in vitro approaches. ddb 64-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11259321-1 2001 Recently we reported that resveratrol (trans-3,4",5-trihydroxystilbene) showed selective inhibition of recombinant human cytochrome P450 (P450) 1A1 in a concentration-dependent manner. Resveratrol 26-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-147 11259321-1 2001 Recently we reported that resveratrol (trans-3,4",5-trihydroxystilbene) showed selective inhibition of recombinant human cytochrome P450 (P450) 1A1 in a concentration-dependent manner. trans-3,4",5-trihydroxystilbene 39-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-147 11392178-3 2001 Phenytoin, an anti-convulsant agent, can induce the Cytochrome P450 enzyme in the liver, which metabolizes Tacrolimus. Tacrolimus 107-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 11270913-3 2001 Because fluvoxamine is an inhibitor of several cytochrome P450 (CYP) enzymes, the authors studied the biotransformation of melatonin and the effects of fluvoxamine on the metabolism of melatonin in vitro using human liver microsomes and recombinant human CYP isoenzymes. Fluvoxamine 8-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 11270913-3 2001 Because fluvoxamine is an inhibitor of several cytochrome P450 (CYP) enzymes, the authors studied the biotransformation of melatonin and the effects of fluvoxamine on the metabolism of melatonin in vitro using human liver microsomes and recombinant human CYP isoenzymes. Fluvoxamine 8-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 11270913-3 2001 Because fluvoxamine is an inhibitor of several cytochrome P450 (CYP) enzymes, the authors studied the biotransformation of melatonin and the effects of fluvoxamine on the metabolism of melatonin in vitro using human liver microsomes and recombinant human CYP isoenzymes. Melatonin 185-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 11392178-3 2001 Phenytoin, an anti-convulsant agent, can induce the Cytochrome P450 enzyme in the liver, which metabolizes Tacrolimus. Phenytoin 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 11411376-7 2001 Some recent reports about interactions (involving hepatic cytochrome P450 enzymes) between dextromethorphan and other drugs are also noteworthy. Dextromethorphan 91-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 11742579-3 2001 The substrates for measuring the activity of individual CYP in vitro included phenacetin, tolbutamide, debrisoquine, and omeprazole. Phenacetin 78-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 11304902-2 2001 Alosetron is extensively metabolized by multiple cytochrome P450 (CYP) enzymes, including CYP 2C9 and 3A4. alosetron 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 11304902-2 2001 Alosetron is extensively metabolized by multiple cytochrome P450 (CYP) enzymes, including CYP 2C9 and 3A4. alosetron 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 11304903-2 2001 Alosetron is extensively metabolized by multiple cytochrome P450 (CYP) enzymes, including CYP2C9 and CYP3A4. alosetron 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 11304903-2 2001 Alosetron is extensively metabolized by multiple cytochrome P450 (CYP) enzymes, including CYP2C9 and CYP3A4. alosetron 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 11274000-1 2001 BACKGROUND: Both a vascular endothelial cytochrome P450 (CYP450) product of arachidonic acid metabolism and the potassium ion (K(+)) have been identified as endothelium-derived hyperpolarizing factors (EDHFs) in animal vascular tissues. Arachidonic Acid 76-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 11274000-1 2001 BACKGROUND: Both a vascular endothelial cytochrome P450 (CYP450) product of arachidonic acid metabolism and the potassium ion (K(+)) have been identified as endothelium-derived hyperpolarizing factors (EDHFs) in animal vascular tissues. Arachidonic Acid 76-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-63 11274000-1 2001 BACKGROUND: Both a vascular endothelial cytochrome P450 (CYP450) product of arachidonic acid metabolism and the potassium ion (K(+)) have been identified as endothelium-derived hyperpolarizing factors (EDHFs) in animal vascular tissues. edhfs 202-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 11274000-1 2001 BACKGROUND: Both a vascular endothelial cytochrome P450 (CYP450) product of arachidonic acid metabolism and the potassium ion (K(+)) have been identified as endothelium-derived hyperpolarizing factors (EDHFs) in animal vascular tissues. edhfs 202-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-63 11274000-9 2001 EDHF-mediated vasorelaxation, however, was sensitive to the phospholipase A(2) inhibitor oleyloxyethyl phosphorylcholine and the CYP450 inhibitor ketoconazole. edhf 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-135 11274000-9 2001 EDHF-mediated vasorelaxation, however, was sensitive to the phospholipase A(2) inhibitor oleyloxyethyl phosphorylcholine and the CYP450 inhibitor ketoconazole. Ketoconazole 146-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-135 11274000-11 2001 A product of phospholipase A(2)/CYP450-dependent metabolism of arachidonic acid and not K(+) is the likely identity of EDHF in human subcutaneous resistance arteries. Arachidonic Acid 63-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-38 11274000-11 2001 A product of phospholipase A(2)/CYP450-dependent metabolism of arachidonic acid and not K(+) is the likely identity of EDHF in human subcutaneous resistance arteries. edhf 119-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-38 11237731-1 2001 CYP2A6 is known as a major cytochrome P450 (CYP) responsible for the oxidation of nicotine and coumarin in humans. Nicotine 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 11237731-1 2001 CYP2A6 is known as a major cytochrome P450 (CYP) responsible for the oxidation of nicotine and coumarin in humans. Nicotine 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 11237731-1 2001 CYP2A6 is known as a major cytochrome P450 (CYP) responsible for the oxidation of nicotine and coumarin in humans. coumarin 95-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 11237731-1 2001 CYP2A6 is known as a major cytochrome P450 (CYP) responsible for the oxidation of nicotine and coumarin in humans. coumarin 95-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 11383698-1 2001 Unsaturated fatty acids with triple bonds are used as inhibitors of unsaturated fatty acid metabolism or cytochrome P450 reactions because they are believed to be chemically inert. Fatty Acids, Unsaturated 0-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 11742579-3 2001 The substrates for measuring the activity of individual CYP in vitro included phenacetin, tolbutamide, debrisoquine, and omeprazole. Tolbutamide 90-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 11742579-3 2001 The substrates for measuring the activity of individual CYP in vitro included phenacetin, tolbutamide, debrisoquine, and omeprazole. Debrisoquin 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 11742579-3 2001 The substrates for measuring the activity of individual CYP in vitro included phenacetin, tolbutamide, debrisoquine, and omeprazole. Omeprazole 121-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 11357344-4 2001 EHF EMR was also inefficient with cells pretreated with proadifen, an inhibitor of epoxygenase (cytochrome P-450). Proadifen 56-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 11240973-1 2001 BACKGROUND AND OBJECTIVES: Fluvoxamine, a selective serotonin reuptake inhibitor, is known to inhibit several hepatic cytochrome P450 (CYP) isozymes, in particular CYP1A2. Fluvoxamine 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 11298070-0 2001 Identification of the cytochrome P450 enzymes involved in the N-demethylation of sildenafil. Nitrogen 62-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 11298070-0 2001 Identification of the cytochrome P450 enzymes involved in the N-demethylation of sildenafil. Sildenafil Citrate 81-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 11298070-1 2001 AIMS: To characterize the cytochrome P450 (CYP) enzymes responsible for the N-demethylation of sildenafil to its main metabolite, UK-103 320, to investigate the potential inhibitory effects of sildenafil on CYP enzymes and to evaluate the potential of selected drugs to affect sildenafil metabolism. Sildenafil Citrate 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 11298070-1 2001 AIMS: To characterize the cytochrome P450 (CYP) enzymes responsible for the N-demethylation of sildenafil to its main metabolite, UK-103 320, to investigate the potential inhibitory effects of sildenafil on CYP enzymes and to evaluate the potential of selected drugs to affect sildenafil metabolism. Sildenafil Citrate 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 11298070-1 2001 AIMS: To characterize the cytochrome P450 (CYP) enzymes responsible for the N-demethylation of sildenafil to its main metabolite, UK-103 320, to investigate the potential inhibitory effects of sildenafil on CYP enzymes and to evaluate the potential of selected drugs to affect sildenafil metabolism. Sildenafil Citrate 193-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 11298070-1 2001 AIMS: To characterize the cytochrome P450 (CYP) enzymes responsible for the N-demethylation of sildenafil to its main metabolite, UK-103 320, to investigate the potential inhibitory effects of sildenafil on CYP enzymes and to evaluate the potential of selected drugs to affect sildenafil metabolism. Sildenafil Citrate 193-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-46 11298070-5 2001 RESULTS: Sildenafil conversion was found to be mediated by at least two CYP enzymes, for which the mean kinetic parameters were Km1 = 6(+/-3 microM), Km2 = 81(+/-45 microM), Vmax1 = 22(+/-9 pmol) and Vmax2 = 138(+/-77 pmol) UK-103 320 formed min(-1) mg(-1). Sildenafil Citrate 9-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 11298076-1 2001 AIMS: To determine the cytochrome P450 (CYP) isoforms involved in the oxidation of propofol by human liver microsomes. Propofol 83-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 11298076-1 2001 AIMS: To determine the cytochrome P450 (CYP) isoforms involved in the oxidation of propofol by human liver microsomes. Propofol 83-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 11298076-2 2001 METHODS: The rate constant calculated from the disappearance of propofol in an incubation mixture with human liver microsomes and recombinant human CYP isoforms was used as a measure of the rate of metabolism of propofol. Propofol 212-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 11298076-5 2001 The rate constants of propofol by microsomes were significantly correlated with S-mephenytoin N-demethylation, a marker of CYP2B6 (r = 0.93, P < 0.0001), but not with the metabolic activities of other CYP isoform-selective substrates. Propofol 22-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-126 11298076-6 2001 Of the chemical inhibitors of CYP isoforms tested, orphenadrine, a CYP2B6 inhibitor, reduced the rate constant of propofol by liver microsomes by 38% (P < 0.05), while other CYP isoform-selective inhibitors had no effects. Orphenadrine 51-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 11298076-6 2001 Of the chemical inhibitors of CYP isoforms tested, orphenadrine, a CYP2B6 inhibitor, reduced the rate constant of propofol by liver microsomes by 38% (P < 0.05), while other CYP isoform-selective inhibitors had no effects. Orphenadrine 51-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11298076-6 2001 Of the chemical inhibitors of CYP isoforms tested, orphenadrine, a CYP2B6 inhibitor, reduced the rate constant of propofol by liver microsomes by 38% (P < 0.05), while other CYP isoform-selective inhibitors had no effects. Propofol 114-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 11298076-6 2001 Of the chemical inhibitors of CYP isoforms tested, orphenadrine, a CYP2B6 inhibitor, reduced the rate constant of propofol by liver microsomes by 38% (P < 0.05), while other CYP isoform-selective inhibitors had no effects. Propofol 114-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11298076-7 2001 Of the recombinant CYP isoforms screened, CYP2B6 produced the highest rate constant for propofol metabolism (197 nmol min-1 nmol P450-1). Propofol 88-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11240973-1 2001 BACKGROUND AND OBJECTIVES: Fluvoxamine, a selective serotonin reuptake inhibitor, is known to inhibit several hepatic cytochrome P450 (CYP) isozymes, in particular CYP1A2. Fluvoxamine 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 11139472-2 2001 As some CYP enzymes are reported to generate reactive oxygen species (ROS), we hypothesized that the coronary EDHF synthase may modulate vascular homeostasis by the simultaneous production of ROS and epoxyeicosatrienoic acids. Reactive Oxygen Species 192-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-11 11317702-0 2001 Study on the cytochrome P-450- and glutathione-dependent biotransformation of trichloroethylene in humans. Trichloroethylene 78-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-30 11263680-1 2001 Mammalian NADPH-ferredoxin reductase (EC 1.18.1.2) functions in the mitochondrial electron transport chain for cytochrome P-450-dependent steroid hydroxylation. Steroids 138-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 11241591-0 2001 The High-Valent Compound of Cytochrome P450: The Nature of the Fe-S Bond and the Role of the Thiolate Ligand as an Internal Electron Donor This research was sponsored in part by the Israeli Science Foundation (ISF) and the Binational German-Israeli Foundation (GIF). Iron 63-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 11241591-0 2001 The High-Valent Compound of Cytochrome P450: The Nature of the Fe-S Bond and the Role of the Thiolate Ligand as an Internal Electron Donor This research was sponsored in part by the Israeli Science Foundation (ISF) and the Binational German-Israeli Foundation (GIF). Sulfur 66-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 11241591-0 2001 The High-Valent Compound of Cytochrome P450: The Nature of the Fe-S Bond and the Role of the Thiolate Ligand as an Internal Electron Donor This research was sponsored in part by the Israeli Science Foundation (ISF) and the Binational German-Israeli Foundation (GIF). thiolate 93-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 11159809-0 2001 Stereochemical aspects of vinylcyclohexene bioactivation in rodent hepatic microsomes and purified human cytochrome P450 enzyme systems. 4-vinylcyclohexene 26-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 11139472-2 2001 As some CYP enzymes are reported to generate reactive oxygen species (ROS), we hypothesized that the coronary EDHF synthase may modulate vascular homeostasis by the simultaneous production of ROS and epoxyeicosatrienoic acids. Reactive Oxygen Species 45-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-11 11139472-2 2001 As some CYP enzymes are reported to generate reactive oxygen species (ROS), we hypothesized that the coronary EDHF synthase may modulate vascular homeostasis by the simultaneous production of ROS and epoxyeicosatrienoic acids. Reactive Oxygen Species 70-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-11 11235806-1 2001 2-(Allylthio)pyrazine derivatives were designed as a novel cancer chemopreventive agent that functions through selective inhibtion of cytochrome P-450 and induction of phase II enzymes involved in the detoxification of carcinogens. 2-(allylthio)pyrazine 0-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 11240372-9 2001 These results suggested that nNOS was more stable as to exposure to sodium cholate, mercury chloride or urea in comparison to microsomal cytochrome P-450, which may be due to the different heme environment and protein structure. Heme 189-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-153 11213066-7 2001 CONCLUSIONS: Patients taking SJW concomitantly with CSA or other medications whose absorption and metabolism are mediated by cytochrome P-450 and/or P-glycoprotein should require close monitoring. Cyclosporine 52-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 11139472-2 2001 As some CYP enzymes are reported to generate reactive oxygen species (ROS), we hypothesized that the coronary EDHF synthase may modulate vascular homeostasis by the simultaneous production of ROS and epoxyeicosatrienoic acids. epoxyeicosatrienoic acids 200-225 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-11 11139472-3 2001 In bradykinin-stimulated coronary arteries, antisense oligonucleotides against CYP 2C almost abolished EDHF-mediated responses but potentiated nitric oxide (NO)-mediated relaxation. Oligonucleotides 54-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 11139472-3 2001 In bradykinin-stimulated coronary arteries, antisense oligonucleotides against CYP 2C almost abolished EDHF-mediated responses but potentiated nitric oxide (NO)-mediated relaxation. edhf 103-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 11475198-4 2001 RESULT: More than 50% of the overall CYP metabolism is mediated through the isoenzyme CYP3A4, which is the main elimination route of simvastatin, lovastatin and atorvastatin. Simvastatin 133-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 11139472-3 2001 In bradykinin-stimulated coronary arteries, antisense oligonucleotides against CYP 2C almost abolished EDHF-mediated responses but potentiated nitric oxide (NO)-mediated relaxation. Nitric Oxide 143-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 11475198-4 2001 RESULT: More than 50% of the overall CYP metabolism is mediated through the isoenzyme CYP3A4, which is the main elimination route of simvastatin, lovastatin and atorvastatin. Lovastatin 146-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 11139472-5 2001 CYP activity and O(2-) production, determined in microsomes prepared from cells overexpressing CYP 2C9, were almost completely inhibited by sulfaphenazole. Sulfaphenazole 140-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 11475198-4 2001 RESULT: More than 50% of the overall CYP metabolism is mediated through the isoenzyme CYP3A4, which is the main elimination route of simvastatin, lovastatin and atorvastatin. Atorvastatin 161-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 11139472-11 2001 These results suggest that a CYP isozyme homologous to CYP 2C9 is a physiologically relevant generator of ROS in coronary endothelial cells and modulates both vascular tone and homeostasis. Reactive Oxygen Species 106-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 11236564-3 2001 It is obvious that microsomal monooxygenase uses the energy of NADPH oxidation for the activation of the terminal agent--cytochrome P-450. NADP 63-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 11730569-1 2001 AIM: The present study was designed to define the kinetic behavior of fluoxetine N-demethylation in human liver microsomes and to identify the isoforms of cytochrome P-450 (CYP) involved in this metabolic pathway. fluoxetine n 70-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 11730569-1 2001 AIM: The present study was designed to define the kinetic behavior of fluoxetine N-demethylation in human liver microsomes and to identify the isoforms of cytochrome P-450 (CYP) involved in this metabolic pathway. fluoxetine n 70-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-176 11730569-10 2001 CONCLUSION: Cytochrome P-450 CYP2C9 is likely to be a major CYP isoform catalyzing fluoxetine N-demethylation in human liver microsomes at a substrate concentration close to the therapeutic level, while polymorphic CYP2C19 may play a more important role in this metabolic pathway at high substrate concentration. fluoxetine n 83-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 11730569-10 2001 CONCLUSION: Cytochrome P-450 CYP2C9 is likely to be a major CYP isoform catalyzing fluoxetine N-demethylation in human liver microsomes at a substrate concentration close to the therapeutic level, while polymorphic CYP2C19 may play a more important role in this metabolic pathway at high substrate concentration. fluoxetine n 83-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-32 12174676-0 2001 Cytochrome P450 polymorphisms as risk factors for steroid hormone-related cancers. Steroids 50-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 12174676-2 2001 Many steroids and environmental carcinogens are subject to cytochrome P450 (P450)-mediated metabolism that generates reactive metabolites and modulates steroid potency, thereby influencing tumor initiation and promotion respectively. Steroids 5-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 12174676-2 2001 Many steroids and environmental carcinogens are subject to cytochrome P450 (P450)-mediated metabolism that generates reactive metabolites and modulates steroid potency, thereby influencing tumor initiation and promotion respectively. Steroids 5-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 11556126-0 2001 Cytochrome P450 enzymes involved in the metabolic pathway of the histamine 2 (H2)-receptor antagonist roxatidine acetate by human liver microsomes. roxatidine acetate 102-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11556126-3 2001 In this study, the Cytochrome P450 (CYP or P450) enzymes which participate in the metabolism of ROX were identified using human liver microsomes and S9 fractions. ROX 96-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 11327199-4 2001 Delavirdine undergoes extensive metabolism by cytochrome P450 (CYP) with little urinary excretion of unchanged drug. Delavirdine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 11523725-3 2001 Pharmacogenetic polymorphism of cytochrome P450 (CYP) may be associated with impaired elimination of warfarin and exaggerated anticoagulatory responses to the drug in certain patients. Warfarin 101-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 11523725-3 2001 Pharmacogenetic polymorphism of cytochrome P450 (CYP) may be associated with impaired elimination of warfarin and exaggerated anticoagulatory responses to the drug in certain patients. Warfarin 101-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 11893055-5 2001 Recent work uses a mimic of the enzyme class cytochrome P-450 to achieve the selective hydroxylations of steroids with complete domination by the geometry of the catalyst-substrate complex. Steroids 105-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 11327199-4 2001 Delavirdine undergoes extensive metabolism by cytochrome P450 (CYP) with little urinary excretion of unchanged drug. Delavirdine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 11327199-6 2001 Concomitant use of CYP inducers, such as rifampicin (rifampin), rifabutin, phenytoin, phenobarbital or carbamazepine, should be avoided since delavirdine plasma concentrations are significantly lowered. Rifampin 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11524025-4 2001 PAHs have been shown to induce 3 hepatic cytochrome P450 (CYP) isozymes, primarily CYP1A1, 1A2 and 2E1. Polycyclic Aromatic Hydrocarbons 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 11327199-6 2001 Concomitant use of CYP inducers, such as rifampicin (rifampin), rifabutin, phenytoin, phenobarbital or carbamazepine, should be avoided since delavirdine plasma concentrations are significantly lowered. Rifampin 53-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11524025-4 2001 PAHs have been shown to induce 3 hepatic cytochrome P450 (CYP) isozymes, primarily CYP1A1, 1A2 and 2E1. Polycyclic Aromatic Hydrocarbons 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 11327199-6 2001 Concomitant use of CYP inducers, such as rifampicin (rifampin), rifabutin, phenytoin, phenobarbital or carbamazepine, should be avoided since delavirdine plasma concentrations are significantly lowered. Rifabutin 64-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11327199-6 2001 Concomitant use of CYP inducers, such as rifampicin (rifampin), rifabutin, phenytoin, phenobarbital or carbamazepine, should be avoided since delavirdine plasma concentrations are significantly lowered. Phenytoin 75-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11327199-6 2001 Concomitant use of CYP inducers, such as rifampicin (rifampin), rifabutin, phenytoin, phenobarbital or carbamazepine, should be avoided since delavirdine plasma concentrations are significantly lowered. Carbamazepine 103-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11327199-6 2001 Concomitant use of CYP inducers, such as rifampicin (rifampin), rifabutin, phenytoin, phenobarbital or carbamazepine, should be avoided since delavirdine plasma concentrations are significantly lowered. Delavirdine 142-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11113705-1 2000 The genotoxicities of four samples of diesel exhaust particle (DEP) extracts (DEPE) and nine nitroarenes found in DEPE were investigated after activation catalyzed by human cytochrome P450 (P450) family 1 enzymes co-expressed with NADPH-cytochrome P450 reductase (NPR) in Escherichia coli membranes. nitroarenes 93-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 173-204 11195057-0 2001 Cytochrome P450 metabolites of arachidonic acid in the control of renal function. Arachidonic Acid 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 11195057-1 2001 Recent studies indicate that arachidonic acid is primarily metabolized by cytochrome P450 enzymes of the 4A and 2C families in the kidney to 20-hydroxyeicosatetraenoic acid (HETE), epoxyeicosatrienoic acids (EETs) and dihydroxyeicosatrienoic acids. Arachidonic Acid 29-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 11195057-1 2001 Recent studies indicate that arachidonic acid is primarily metabolized by cytochrome P450 enzymes of the 4A and 2C families in the kidney to 20-hydroxyeicosatetraenoic acid (HETE), epoxyeicosatrienoic acids (EETs) and dihydroxyeicosatrienoic acids. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 141-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 11195057-1 2001 Recent studies indicate that arachidonic acid is primarily metabolized by cytochrome P450 enzymes of the 4A and 2C families in the kidney to 20-hydroxyeicosatetraenoic acid (HETE), epoxyeicosatrienoic acids (EETs) and dihydroxyeicosatrienoic acids. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 174-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 11195057-1 2001 Recent studies indicate that arachidonic acid is primarily metabolized by cytochrome P450 enzymes of the 4A and 2C families in the kidney to 20-hydroxyeicosatetraenoic acid (HETE), epoxyeicosatrienoic acids (EETs) and dihydroxyeicosatrienoic acids. epoxyeicosatrienoic acids 181-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 11195057-1 2001 Recent studies indicate that arachidonic acid is primarily metabolized by cytochrome P450 enzymes of the 4A and 2C families in the kidney to 20-hydroxyeicosatetraenoic acid (HETE), epoxyeicosatrienoic acids (EETs) and dihydroxyeicosatrienoic acids. eets 208-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 11195057-1 2001 Recent studies indicate that arachidonic acid is primarily metabolized by cytochrome P450 enzymes of the 4A and 2C families in the kidney to 20-hydroxyeicosatetraenoic acid (HETE), epoxyeicosatrienoic acids (EETs) and dihydroxyeicosatrienoic acids. dihydroxyeicosatrienoic acids 218-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 11774365-3 2001 The expression levels of CYP holo-protein in the genetically engineered S. typhimurium YG7108 cells, determined by carbon monoxide (CO) difference spectra, ranged from 62 nmol/L culture for CYP2C19 to 169 nmol/L culture for CYP3A4. Carbon Monoxide 115-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 11774365-5 2001 Each form of CYP expressed in the S. typhimurium YG7108 cells catalyzed the oxidation of a representative substrate at an efficient rate. yg7108 49-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 11774366-8 2001 Thus, the addition of a pyridine ring to NPYR or NPIP altered the forms of CYP primarily responsible for mutagenic activation. pyridine 24-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 21336918-6 2001 Arachidonic acid can also be metabolized by specific cytochrome P(450) enzymes to regioisomeric epoxides and stereo specific hydro xyeicosatetraenoic (HETE) acids (6). Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-70 21336918-6 2001 Arachidonic acid can also be metabolized by specific cytochrome P(450) enzymes to regioisomeric epoxides and stereo specific hydro xyeicosatetraenoic (HETE) acids (6). Epoxy Compounds 96-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-70 21336918-6 2001 Arachidonic acid can also be metabolized by specific cytochrome P(450) enzymes to regioisomeric epoxides and stereo specific hydro xyeicosatetraenoic (HETE) acids (6). Hydroxyeicosatetraenoic Acids 125-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-70 11195057-11 2001 The renal production of cytochrome P450 metabolites of arachidonic acid is altered in hypertension, diabetes, toxemia of pregnancy, and hepatorenal syndrome. Arachidonic Acid 55-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 11195057-12 2001 Given the importance of cytochrome P450 metabolites of arachidonic acid in the control of renal function, it is likely that changes in this system contribute to the abnormalities in renal function that are associated with many of these conditions. Arachidonic Acid 55-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 12-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 11192140-3 2000 Both citalopram and theophylline are metabolized by cytochrome P450 (CYP) isozymes. Citalopram 5-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 12-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 11192140-3 2000 Both citalopram and theophylline are metabolized by cytochrome P450 (CYP) isozymes. Citalopram 5-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-80 11192140-3 2000 Both citalopram and theophylline are metabolized by cytochrome P450 (CYP) isozymes. Theophylline 20-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 11465044-9 2000 If the latter is stable it may undergo oxidation by cytochrome P-450 to form the lactam. Lactams 81-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 11180018-1 2000 BACKGROUND: Rifampin (rifampicin) is a potent inducer of several cytochrome P450 (CYP) enzymes, including CYP3A4. Rifampin 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 11192140-3 2000 Both citalopram and theophylline are metabolized by cytochrome P450 (CYP) isozymes. Theophylline 20-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 11095582-0 2000 Identification of the human liver cytochrome P450 isoenzyme responsible for the 6-methylhydroxylation of the novel anticancer drug 5,6-dimethylxanthenone-4-acetic acid. vadimezan 131-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 11095582-6 2000 The involvement of CYP1A2 in DMXAA metabolism by human livers was demonstrated by the following: 1) the potent inhibition of DMXAA metabolism by furafylline (k(inact) = 0.23 +/- 0.04 min(-1), K"(app) = 15.6 +/- 6.7 microM) and alpha-naphthoflavone (K(i) = 0.036 microM), but not by cimetidine, ketoconazole, tolbutamide, quinidine, chlorzoxazone, diethyldithiocarbamate, troleandomycin, and sulfaphenazole; 2) when incubated with human lymphoblastoid cell microsomes containing cDNA-expressed CYP isoenzymes, DMXAA was metabolized only by CYP1A2, with an apparent K(m) of 6.2 +/- 1.5 microM and V(max) of 0.014 +/- 0.001 nmol/min/mg, but not by CYP2A6, CYP2B6, CYP2C9 (Arg(144)), CYP2C19, CYP2D6 (Val(374)), CYP2E1, and CYP3A4; 3) a significant correlation (r = 0.90; P <.001) between 6-methylhydroxylation of DMXAA and 7-ethoxyresorufin O-deethylation; and 4) a significant correlation (r = 0.75; P <.01) between the CYP1A protein level determined by Western blots and DMXAA 6-methylhydroxylation. furafylline 145-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11095582-1 2000 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) isoenzyme involved in the 6-methylhydroxylation of 5, 6-dimethylxanthenone-4-acetic acid (DMXAA) by using a human liver library (n = 14). vadimezan 129-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 11095582-1 2000 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) isoenzyme involved in the 6-methylhydroxylation of 5, 6-dimethylxanthenone-4-acetic acid (DMXAA) by using a human liver library (n = 14). vadimezan 129-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 11095582-1 2000 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) isoenzyme involved in the 6-methylhydroxylation of 5, 6-dimethylxanthenone-4-acetic acid (DMXAA) by using a human liver library (n = 14). vadimezan 168-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 11095582-1 2000 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) isoenzyme involved in the 6-methylhydroxylation of 5, 6-dimethylxanthenone-4-acetic acid (DMXAA) by using a human liver library (n = 14). vadimezan 168-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 11095582-3 2000 The metabolite formed in human liver microsomes and by cDNA-expressed CYP isoform was identified by liquid chromatography mass spectrometry as 6-OH-MXAA. 6-oh-mxaa 143-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 11095582-6 2000 The involvement of CYP1A2 in DMXAA metabolism by human livers was demonstrated by the following: 1) the potent inhibition of DMXAA metabolism by furafylline (k(inact) = 0.23 +/- 0.04 min(-1), K"(app) = 15.6 +/- 6.7 microM) and alpha-naphthoflavone (K(i) = 0.036 microM), but not by cimetidine, ketoconazole, tolbutamide, quinidine, chlorzoxazone, diethyldithiocarbamate, troleandomycin, and sulfaphenazole; 2) when incubated with human lymphoblastoid cell microsomes containing cDNA-expressed CYP isoenzymes, DMXAA was metabolized only by CYP1A2, with an apparent K(m) of 6.2 +/- 1.5 microM and V(max) of 0.014 +/- 0.001 nmol/min/mg, but not by CYP2A6, CYP2B6, CYP2C9 (Arg(144)), CYP2C19, CYP2D6 (Val(374)), CYP2E1, and CYP3A4; 3) a significant correlation (r = 0.90; P <.001) between 6-methylhydroxylation of DMXAA and 7-ethoxyresorufin O-deethylation; and 4) a significant correlation (r = 0.75; P <.01) between the CYP1A protein level determined by Western blots and DMXAA 6-methylhydroxylation. vadimezan 29-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11095582-6 2000 The involvement of CYP1A2 in DMXAA metabolism by human livers was demonstrated by the following: 1) the potent inhibition of DMXAA metabolism by furafylline (k(inact) = 0.23 +/- 0.04 min(-1), K"(app) = 15.6 +/- 6.7 microM) and alpha-naphthoflavone (K(i) = 0.036 microM), but not by cimetidine, ketoconazole, tolbutamide, quinidine, chlorzoxazone, diethyldithiocarbamate, troleandomycin, and sulfaphenazole; 2) when incubated with human lymphoblastoid cell microsomes containing cDNA-expressed CYP isoenzymes, DMXAA was metabolized only by CYP1A2, with an apparent K(m) of 6.2 +/- 1.5 microM and V(max) of 0.014 +/- 0.001 nmol/min/mg, but not by CYP2A6, CYP2B6, CYP2C9 (Arg(144)), CYP2C19, CYP2D6 (Val(374)), CYP2E1, and CYP3A4; 3) a significant correlation (r = 0.90; P <.001) between 6-methylhydroxylation of DMXAA and 7-ethoxyresorufin O-deethylation; and 4) a significant correlation (r = 0.75; P <.01) between the CYP1A protein level determined by Western blots and DMXAA 6-methylhydroxylation. vadimezan 125-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 11102737-15 2000 The taxanes paclitaxel and docetaxel are considered excellent substrate drugs to test the concept that by inhibition of P-gp in the gut wall and CYP activity in gut wall and/or liver low oral bioavailability can be increased substantially. Paclitaxel 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 11145063-9 2000 The method was applied to the determination of the rate and enantioselectivity of the cytochrome P-450 dependent oxidation of styrene to SO enantiomers in human liver microsomes. Styrene 126-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 11102737-15 2000 The taxanes paclitaxel and docetaxel are considered excellent substrate drugs to test the concept that by inhibition of P-gp in the gut wall and CYP activity in gut wall and/or liver low oral bioavailability can be increased substantially. Docetaxel 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 11061988-2 2000 We employed well-characterized inhibitors of specific cytochrome P-450 (CYP) enzymes to determine which form was likely involved in tocopherol side chain oxidation. Tocopherols 132-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. Ifosfamide 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. Ifosfamide 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. chloroacetaldehyde 138-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. chloroacetaldehyde 138-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. chloroacetaldehyde 193-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. chloroacetaldehyde 193-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. Ifosfamide 217-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. Ifosfamide 217-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. Cyclophosphamide 238-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-65 11132240-4 2000 We hypothesized that ifosfamide is metabolized by cytochrome P450 (CYP) enzymes located in the renal tubular cell to the toxic metabolite chloroacetaldehyde; and, that the higher production of chloroacetaldehyde from ifosfamide than from cyclophosphamide explains the clinical differences in nephrotoxicity. Cyclophosphamide 238-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 11144010-5 2000 Thyroxin is a potent activator of the cytochrome P-450- CYP 3A enzyme system, which is crucial for tacrolimus metabolism. Tacrolimus 99-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 11144010-8 2000 In conclusion, it is important to consider thyroid function when prescribing medications with a narrow therapeutic range, which are metabolized by the cytochrome P-450 system such as tacrolimus, and the possible devastating effect of impaired drug metabolism during hypothyroidism. Tacrolimus 183-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 11084219-1 2000 The present study was designed to determine the effect of venlafaxine on imipramine metabolism in an attempt to elucidate the potential for cytochrome P450 drug-drug interactions with venlafaxine. Venlafaxine Hydrochloride 184-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-155 29711670-0 2000 The High-Valent Compound of Cytochrome P450: The Nature of the Fe-S Bond and the Role of the Thiolate Ligand as an Internal Electron Donor. Iron 63-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 29711670-0 2000 The High-Valent Compound of Cytochrome P450: The Nature of the Fe-S Bond and the Role of the Thiolate Ligand as an Internal Electron Donor. Sulfur 66-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 29711670-0 2000 The High-Valent Compound of Cytochrome P450: The Nature of the Fe-S Bond and the Role of the Thiolate Ligand as an Internal Electron Donor. thiolate 93-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 11061988-2 2000 We employed well-characterized inhibitors of specific cytochrome P-450 (CYP) enzymes to determine which form was likely involved in tocopherol side chain oxidation. Tocopherols 132-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 11113932-0 2000 Stereoselective determination of propafenone enantiomers in transgenic Chinese hamster CHL cells expressing human cytochrome P450. Propafenone 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 11113932-1 2000 An enantioselective assay for S(+)- and R(-)-propafenone in transgenic Chinese hamster CHL cells expressing human cytochrome P450 was developed. s(+)- and r(-)-propafenone 30-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-129 11113932-8 2000 The method allowed study of the depletion of S(+)- and R(-)-propafenone in transgenic Chinese hamster CHL cells expressing human cytochrome P450. Sulfur 45-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-144 11113932-8 2000 The method allowed study of the depletion of S(+)- and R(-)-propafenone in transgenic Chinese hamster CHL cells expressing human cytochrome P450. Propafenone 55-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-144 11038149-3 2000 We previously reported that the human cytochrome P450 (P450), 2A6, efficiently and specifically catalyzed NNN 5"-hydroxylation. nnn 5" 106-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-65 11106261-2 2000 It is bioactivated to 5-FU via 5"-hydroxylation mediated by cytochrome P-450 (CYP). Fluorouracil 22-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 11106261-2 2000 It is bioactivated to 5-FU via 5"-hydroxylation mediated by cytochrome P-450 (CYP). Fluorouracil 22-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-81 11038157-1 2000 Human cytochrome P450 (CYP) isoforms involved in amiodarone N-deethylation were identified, and the relative contributions of these CYP isoforms were evaluated in different human liver microsomes. Amiodarone 49-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 11038157-7 2000 However, there was a large interindividual variability in the contribution of each CYP isoform to amiodarone N-deethylase activity in human liver; the relevance of these enzymes would be dependent on the content of the respective isoforms and on the amiodarone concentration in the liver. Amiodarone 98-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 11038157-7 2000 However, there was a large interindividual variability in the contribution of each CYP isoform to amiodarone N-deethylase activity in human liver; the relevance of these enzymes would be dependent on the content of the respective isoforms and on the amiodarone concentration in the liver. Amiodarone 250-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 11038157-1 2000 Human cytochrome P450 (CYP) isoforms involved in amiodarone N-deethylation were identified, and the relative contributions of these CYP isoforms were evaluated in different human liver microsomes. Amiodarone 49-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 11038154-3 2000 The objective of the study was to establish the cytochrome P450 (CYP) enzymes catalyzing the hydroxylations of the acenocoumarol enantiomers. Acenocoumarol 115-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 11038157-2 2000 The mean K(M) and V(max) values of amiodarone N-deethylation in microsomes from six human livers were 31.6 +/- 7.5 microM and 1.2 +/- 0.7 pmol/min/pmol of CYP, respectively. Amiodarone 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-158 11038154-3 2000 The objective of the study was to establish the cytochrome P450 (CYP) enzymes catalyzing the hydroxylations of the acenocoumarol enantiomers. Acenocoumarol 115-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 11038157-4 2000 Of 15 recombinant human CYP enzymes (19 preparations), CYP1A1, CYP3A4, CYP1A2, CYP2D6, CYP2C8, and CYP2C19 catalyzed amiodarone N-deethylation. amiodarone n 117-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 10997941-8 2000 Similar losses of CYP enzymes were found when human liver slices were cultured in supplemented Williams" medium E for 72 h, except that CYP2E1 apoprotein levels were better maintained. williams" medium e 95-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 11038166-1 2000 In an in vitro study, we compared the cytochrome P450 (CYP)-dependent metabolism and drug interactions of the acid and lactone forms of the 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitor atorvastatin. Lactones 119-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 11038166-1 2000 In an in vitro study, we compared the cytochrome P450 (CYP)-dependent metabolism and drug interactions of the acid and lactone forms of the 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitor atorvastatin. Lactones 119-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 11038166-1 2000 In an in vitro study, we compared the cytochrome P450 (CYP)-dependent metabolism and drug interactions of the acid and lactone forms of the 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitor atorvastatin. Atorvastatin 197-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 11038166-1 2000 In an in vitro study, we compared the cytochrome P450 (CYP)-dependent metabolism and drug interactions of the acid and lactone forms of the 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitor atorvastatin. Atorvastatin 197-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-58 11038166-5 2000 Compared with atorvastatin acid, CYP-dependent metabolism of atorvastatin lactone to its para-hydroxy metabolite was 83-fold higher [formation CL(int) (V(max)/K(m)): lactone 2949 +/- 3511 versus acid 35.5 +/- 48.1 microl. Atorvastatin lactone 61-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 11038166-5 2000 Compared with atorvastatin acid, CYP-dependent metabolism of atorvastatin lactone to its para-hydroxy metabolite was 83-fold higher [formation CL(int) (V(max)/K(m)): lactone 2949 +/- 3511 versus acid 35.5 +/- 48.1 microl. Lactones 74-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 10997945-4 2000 Other cytochrome P450 (CYP) 3A4 inhibitors, cimetidine, erythromycin, and clarithromycin, also inhibited the metabolism of cisapride and mosapride. Cimetidine 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 10997945-4 2000 Other cytochrome P450 (CYP) 3A4 inhibitors, cimetidine, erythromycin, and clarithromycin, also inhibited the metabolism of cisapride and mosapride. Cimetidine 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 10997945-4 2000 Other cytochrome P450 (CYP) 3A4 inhibitors, cimetidine, erythromycin, and clarithromycin, also inhibited the metabolism of cisapride and mosapride. Erythromycin 56-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 10997945-4 2000 Other cytochrome P450 (CYP) 3A4 inhibitors, cimetidine, erythromycin, and clarithromycin, also inhibited the metabolism of cisapride and mosapride. Erythromycin 56-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 10997945-4 2000 Other cytochrome P450 (CYP) 3A4 inhibitors, cimetidine, erythromycin, and clarithromycin, also inhibited the metabolism of cisapride and mosapride. Clarithromycin 74-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 10997945-4 2000 Other cytochrome P450 (CYP) 3A4 inhibitors, cimetidine, erythromycin, and clarithromycin, also inhibited the metabolism of cisapride and mosapride. Cisapride 123-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 10997945-4 2000 Other cytochrome P450 (CYP) 3A4 inhibitors, cimetidine, erythromycin, and clarithromycin, also inhibited the metabolism of cisapride and mosapride. Cisapride 123-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 10997945-4 2000 Other cytochrome P450 (CYP) 3A4 inhibitors, cimetidine, erythromycin, and clarithromycin, also inhibited the metabolism of cisapride and mosapride. mosapride 137-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 10997945-4 2000 Other cytochrome P450 (CYP) 3A4 inhibitors, cimetidine, erythromycin, and clarithromycin, also inhibited the metabolism of cisapride and mosapride. mosapride 137-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 11128045-0 2000 Oxidation of ranitidine by isozymes of flavin-containing monooxygenase and cytochrome P450. Ranitidine 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 11069215-5 2000 H2 antagonists, proton pump inhibitors and prokinetic agents undergo metabolism by the cytochrome P450 (CYP) system present in the liver and gastrointestinal tract. Hydrogen 0-2 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 11069215-5 2000 H2 antagonists, proton pump inhibitors and prokinetic agents undergo metabolism by the cytochrome P450 (CYP) system present in the liver and gastrointestinal tract. Hydrogen 0-2 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-107 11128045-2 2000 N- and S-oxidations of ranitidine were inhibited by metimazole [flavin-containing monooxygenase (FMO) inhibitor] to 96-97% and 71-85%, respectively, and desmethylation of ranitidine was inhibited by SKF525A [cytochrome P450 (CYP) inhibitor] by 71-95%. Ranitidine 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 208-223 11128045-2 2000 N- and S-oxidations of ranitidine were inhibited by metimazole [flavin-containing monooxygenase (FMO) inhibitor] to 96-97% and 71-85%, respectively, and desmethylation of ranitidine was inhibited by SKF525A [cytochrome P450 (CYP) inhibitor] by 71-95%. Ranitidine 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 225-228 11128045-2 2000 N- and S-oxidations of ranitidine were inhibited by metimazole [flavin-containing monooxygenase (FMO) inhibitor] to 96-97% and 71-85%, respectively, and desmethylation of ranitidine was inhibited by SKF525A [cytochrome P450 (CYP) inhibitor] by 71-95%. metimazole 52-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 208-223 11128045-2 2000 N- and S-oxidations of ranitidine were inhibited by metimazole [flavin-containing monooxygenase (FMO) inhibitor] to 96-97% and 71-85%, respectively, and desmethylation of ranitidine was inhibited by SKF525A [cytochrome P450 (CYP) inhibitor] by 71-95%. metimazole 52-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 225-228 11128045-2 2000 N- and S-oxidations of ranitidine were inhibited by metimazole [flavin-containing monooxygenase (FMO) inhibitor] to 96-97% and 71-85%, respectively, and desmethylation of ranitidine was inhibited by SKF525A [cytochrome P450 (CYP) inhibitor] by 71-95%. Ranitidine 171-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 208-223 11128045-2 2000 N- and S-oxidations of ranitidine were inhibited by metimazole [flavin-containing monooxygenase (FMO) inhibitor] to 96-97% and 71-85%, respectively, and desmethylation of ranitidine was inhibited by SKF525A [cytochrome P450 (CYP) inhibitor] by 71-95%. Ranitidine 171-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 225-228 11315104-0 2000 Metabolism of 7-benzyloxy-4-trifluoromethyl-coumarin by human hepatic cytochrome P450 isoforms. 7-benzyloxy-4-trifluoromethylcoumarin 14-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 11315104-2 2000 The metabolism of 7-benzyloxy-4-trifluoromethylcoumarin (BFC) to 7-hydroxy-4-trifluoromethylcoumarin (HFC) was studied in human liver microsomal preparations and in cDNA-expressed human cytochrome P450 (CYP) isoforms. 7-benzyloxy-4-trifluoromethylcoumarin 18-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-201 11315104-2 2000 The metabolism of 7-benzyloxy-4-trifluoromethylcoumarin (BFC) to 7-hydroxy-4-trifluoromethylcoumarin (HFC) was studied in human liver microsomal preparations and in cDNA-expressed human cytochrome P450 (CYP) isoforms. 7-benzyloxy-4-trifluoromethylcoumarin 18-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 203-206 11315104-2 2000 The metabolism of 7-benzyloxy-4-trifluoromethylcoumarin (BFC) to 7-hydroxy-4-trifluoromethylcoumarin (HFC) was studied in human liver microsomal preparations and in cDNA-expressed human cytochrome P450 (CYP) isoforms. 7-benzyloxy-4-trifluoromethylcoumarin 57-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-201 11315104-2 2000 The metabolism of 7-benzyloxy-4-trifluoromethylcoumarin (BFC) to 7-hydroxy-4-trifluoromethylcoumarin (HFC) was studied in human liver microsomal preparations and in cDNA-expressed human cytochrome P450 (CYP) isoforms. 7-benzyloxy-4-trifluoromethylcoumarin 57-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 203-206 11315104-2 2000 The metabolism of 7-benzyloxy-4-trifluoromethylcoumarin (BFC) to 7-hydroxy-4-trifluoromethylcoumarin (HFC) was studied in human liver microsomal preparations and in cDNA-expressed human cytochrome P450 (CYP) isoforms. 7-hydroxy-4-trifluoromethylcoumarin 65-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-201 11315104-10 2000 Using human beta-lymphoblastoid cell microsomes containing cDNA-expressed CYP isoforms, 20 microM BFC was metabolized by CYP1A2 and CYP3A4, with lower rates of metabolism being observed with CYP2C9 and CYP2C19. 7-benzyloxy-4-trifluoromethylcoumarin 98-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 11315104-18 2000 In summary, by correlation analysis, use of cDNA-expressed CYP isoforms, chemical inhibition and inhibitory antibodies, BFC appears metabolized by a number of CYP isoforms in human liver. 7-benzyloxy-4-trifluoromethylcoumarin 120-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 11315104-18 2000 In summary, by correlation analysis, use of cDNA-expressed CYP isoforms, chemical inhibition and inhibitory antibodies, BFC appears metabolized by a number of CYP isoforms in human liver. 7-benzyloxy-4-trifluoromethylcoumarin 120-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-162 11315104-19 2000 BFC metabolism appears to be primarily catalysed by CYP1A2 and CYP3A4, with possibly some contribution by CYP2C9, CYP2C19 and perhaps other CYP isoforms. 7-benzyloxy-4-trifluoromethylcoumarin 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 10924087-1 2000 Epoxyeicosatrienoic acids (EETs) are cytochrome P-450 metabolites of arachidonic acid involved in the regulation of vascular tone. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 11026737-0 2000 Cytochrome P-450 enzymes and FMO3 contribute to the disposition of the antipsychotic drug perazine in vitro. Perazine 90-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 11005703-13 2000 Fluvastatin has several metabolic pathways which involve the CYP enzyme system. Fluvastatin 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 10874126-0 2000 Identification of human cytochrome P450 isoforms that contribute to all-trans-retinoic acid 4-hydroxylation. 2-octenal 72-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 10874126-0 2000 Identification of human cytochrome P450 isoforms that contribute to all-trans-retinoic acid 4-hydroxylation. Tretinoin 78-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 10874126-1 2000 The role of specific human cytochrome P450 (CYP) isoforms in the oxidative metabolism of all-trans-retinoic acid was investigated by studies in human liver microsomes using isoform-specific chemical inhibitors and inhibitory antibodies. Tretinoin 89-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 10874126-1 2000 The role of specific human cytochrome P450 (CYP) isoforms in the oxidative metabolism of all-trans-retinoic acid was investigated by studies in human liver microsomes using isoform-specific chemical inhibitors and inhibitory antibodies. Tretinoin 89-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 10874126-4 2000 Using inhibition studies and correlation analysis, we also concluded that CYP2C8 was the major all-trans-retinoic acid 4-hydroxylating cytochrome P450 in human liver microsomes, though CYP3A4 and, to a lesser extent CYP2C9, also made a contribution. Tretinoin 95-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-150 10924087-1 2000 Epoxyeicosatrienoic acids (EETs) are cytochrome P-450 metabolites of arachidonic acid involved in the regulation of vascular tone. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 10924087-1 2000 Epoxyeicosatrienoic acids (EETs) are cytochrome P-450 metabolites of arachidonic acid involved in the regulation of vascular tone. Arachidonic Acid 69-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 10948089-2 2000 In some vascular beds, this so-called endothelium-derived hyperpolarizing factor (EDHF) displays the characteristics of a cytochrome P450 (CYP)-derived arachidonic acid metabolite, such as an epoxyeicosatrienoic acid. hyperpolarizing factor 58-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 10948089-2 2000 In some vascular beds, this so-called endothelium-derived hyperpolarizing factor (EDHF) displays the characteristics of a cytochrome P450 (CYP)-derived arachidonic acid metabolite, such as an epoxyeicosatrienoic acid. hyperpolarizing factor 58-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 10948089-2 2000 In some vascular beds, this so-called endothelium-derived hyperpolarizing factor (EDHF) displays the characteristics of a cytochrome P450 (CYP)-derived arachidonic acid metabolite, such as an epoxyeicosatrienoic acid. edhf 82-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 10948089-2 2000 In some vascular beds, this so-called endothelium-derived hyperpolarizing factor (EDHF) displays the characteristics of a cytochrome P450 (CYP)-derived arachidonic acid metabolite, such as an epoxyeicosatrienoic acid. edhf 82-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 10948089-2 2000 In some vascular beds, this so-called endothelium-derived hyperpolarizing factor (EDHF) displays the characteristics of a cytochrome P450 (CYP)-derived arachidonic acid metabolite, such as an epoxyeicosatrienoic acid. Arachidonic Acid 152-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 10948089-2 2000 In some vascular beds, this so-called endothelium-derived hyperpolarizing factor (EDHF) displays the characteristics of a cytochrome P450 (CYP)-derived arachidonic acid metabolite, such as an epoxyeicosatrienoic acid. Arachidonic Acid 152-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 10948089-2 2000 In some vascular beds, this so-called endothelium-derived hyperpolarizing factor (EDHF) displays the characteristics of a cytochrome P450 (CYP)-derived arachidonic acid metabolite, such as an epoxyeicosatrienoic acid. epoxyeicosatrienoic acid 192-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-137 10948089-2 2000 In some vascular beds, this so-called endothelium-derived hyperpolarizing factor (EDHF) displays the characteristics of a cytochrome P450 (CYP)-derived arachidonic acid metabolite, such as an epoxyeicosatrienoic acid. epoxyeicosatrienoic acid 192-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 10948089-4 2000 The CYP inducer beta-naphthoflavone and the Ca(2+) antagonist nifedipine significantly increased CYP2C mRNA but did not change the expression of CYP2J or CYP2B. beta-Naphthoflavone 16-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 10948089-9 2000 These results demonstrate that in porcine coronary arteries, the elevated expression of a CYP epoxygenase, homologous to CYP2C8/9, is associated with enhanced EDHF-mediated hyperpolarization in response to bradykinin. edhf 159-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-93 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. bufuralol 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 10923005-0 2000 Cytochrome P450 polymorphisms are associated with reduced warfarin dose. Warfarin 58-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10923005-2 2000 Interpatient variability associated with warfarin therapy may be partly attributable to polymorphisms of the cytochrome P450 (CYP) complex. Warfarin 41-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 10923005-2 2000 Interpatient variability associated with warfarin therapy may be partly attributable to polymorphisms of the cytochrome P450 (CYP) complex. Warfarin 41-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 10923005-3 2000 The purpose of this study was to ascertain the frequency and influence of CYP polymorphisms on warfarin dosing in our patient population. Warfarin 95-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. bufuralol 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. Testosterone 97-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. Testosterone 97-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. Dextromethorphan 111-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. Dextromethorphan 111-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. Phenacetin 129-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. Diclofenac 141-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. Mephenytoin 156-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 11037108-5 2000 Human CYP probe substrates used for characterization of mouse CYP activities included bufuralol, testosterone, dextromethorphan, phenacetin, diclofenac and S-mephenytoin. Mephenytoin 156-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-65 10926165-1 2000 Several porphyrinogenic xenobiotics elicit mechanism-based inactivation of cytochrome P450 (CYP) isozymes, leading to the formation of N-alkylprotoporphyrin IX (N-alkylPP), a potent inhibitor of ferrochelatase, the terminal enzyme in heme biosynthesis. n-alkylprotoporphyrin ix 135-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 10933836-0 2000 Improving the cytochrome P450 enzyme system for electrode-driven biocatalysis of styrene epoxidation. Styrene 81-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 10933836-3 2000 This paper reports several improvements that make the cytochrome P450cam enzyme system more practical for the epoxidation of styrene. Styrene 125-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 10926165-1 2000 Several porphyrinogenic xenobiotics elicit mechanism-based inactivation of cytochrome P450 (CYP) isozymes, leading to the formation of N-alkylprotoporphyrin IX (N-alkylPP), a potent inhibitor of ferrochelatase, the terminal enzyme in heme biosynthesis. n-alkylprotoporphyrin ix 135-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 10926165-1 2000 Several porphyrinogenic xenobiotics elicit mechanism-based inactivation of cytochrome P450 (CYP) isozymes, leading to the formation of N-alkylprotoporphyrin IX (N-alkylPP), a potent inhibitor of ferrochelatase, the terminal enzyme in heme biosynthesis. n-alkylpp 161-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 10926165-1 2000 Several porphyrinogenic xenobiotics elicit mechanism-based inactivation of cytochrome P450 (CYP) isozymes, leading to the formation of N-alkylprotoporphyrin IX (N-alkylPP), a potent inhibitor of ferrochelatase, the terminal enzyme in heme biosynthesis. n-alkylpp 161-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 10926165-1 2000 Several porphyrinogenic xenobiotics elicit mechanism-based inactivation of cytochrome P450 (CYP) isozymes, leading to the formation of N-alkylprotoporphyrin IX (N-alkylPP), a potent inhibitor of ferrochelatase, the terminal enzyme in heme biosynthesis. Heme 234-238 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-90 10926165-1 2000 Several porphyrinogenic xenobiotics elicit mechanism-based inactivation of cytochrome P450 (CYP) isozymes, leading to the formation of N-alkylprotoporphyrin IX (N-alkylPP), a potent inhibitor of ferrochelatase, the terminal enzyme in heme biosynthesis. Heme 234-238 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 10871337-5 2000 Whereas vincristine and vinblastine at a concentration of 100 nM killed more than 90% of the parental cells, more than 50 and 35%, respectively, of cells that coexpressed CYP3A4 and cytochrome P450 (P450) reductase survived these treatments. Vinblastine 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-214 10871299-5 2000 Hyperforin and I3,II8-biapigenin were isolated from the extract, and inhibition constants for the five CYP activities were measured. hyperforin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 10859153-0 2000 Interaction of cisapride with the human cytochrome P450 system: metabolism and inhibition studies. Cisapride 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 10871299-9 2000 Hypericin also demonstrated potent inhibition of several CYP activities. hypericin 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 10871299-5 2000 Hyperforin and I3,II8-biapigenin were isolated from the extract, and inhibition constants for the five CYP activities were measured. biapigenin 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-106 10871299-6 2000 In addition, three other constituents, hypericin, quercetin, and chlorogenic acid, were tested for inhibitory activity toward the CYP enzymes. hypericin 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 10871299-6 2000 In addition, three other constituents, hypericin, quercetin, and chlorogenic acid, were tested for inhibitory activity toward the CYP enzymes. Chlorogenic Acid 65-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-133 10860554-0 2000 Promoter activity and regulation of the CYP4F2 leukotriene B(4) omega-hydroxylase gene by peroxisomal proliferators and retinoic acid in HepG2 cells. Tretinoin 120-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-81 10860554-2 2000 AF221943) encodes a leukotriene B(4) omega-hydroxylase that metabolizes leukotriene B(4) (LTB(4)) to a less potent proinflammatory eicosanoid, 20-OH-LTB(4). Eicosanoids 131-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-54 10860554-2 2000 AF221943) encodes a leukotriene B(4) omega-hydroxylase that metabolizes leukotriene B(4) (LTB(4)) to a less potent proinflammatory eicosanoid, 20-OH-LTB(4). 20-oh-ltb 143-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-54 10901126-0 2000 Metabolic profiling of valproic acid by cDNA-expressed human cytochrome P450 enzymes using negative-ion chemical ionization gas chromatography-mass spectrometry. Valproic Acid 23-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-76 10901126-1 2000 A sensitive negative ion chemical ionization (NCI) gas chromatographic-mass spectrometric (GC-MS) method was modified for the quantitation of valproic acid (VPA) metabolites generated from in vitro cDNA-expressed human microsomal cytochrome P450 incubations. Valproic Acid 157-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 230-245 10852767-0 2000 Synthesis of arachidonic acid-derived lipoxygenase and cytochrome P450 products in the intact human lung vasculature. Arachidonic Acid 13-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 10941006-0 2000 Hydrogen-Bonded Dioxygen Adduct of an Iron Porphyrin with an Alkanethiolate Ligand: An Elaborate Model of Cytochrome P450 This research was supported by the Ministry of Education, Science, and Culture, Japan (Grant-in Aids #08CE2005, 09235225, and 11228207 to Y.N.) Hydrogen 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-121 10852767-1 2000 Lipoxygenase (LO) and cytochrome P450 monooxygenase products of arachidonic acid (AA) have been implicated in a large number of vasoregulatory processes. Arachidonic Acid 64-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 10820139-0 2000 In vitro inhibition and induction of human hepatic cytochrome P450 enzymes by modafinil. Modafinil 78-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-66 10820139-1 2000 The ability of modafinil to affect human hepatic cytochrome P450 (CYP) activities was examined in vitro. Modafinil 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 10820139-3 2000 Modafinil exhibited minimal capacity to inhibit any CYP enzyme, except CYP2C19. Modafinil 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 10820139-1 2000 The ability of modafinil to affect human hepatic cytochrome P450 (CYP) activities was examined in vitro. Modafinil 15-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 10820139-6 2000 The potential for induction of CYP activity was evaluated by exposing primary cultures of human hepatocytes to modafinil (10-300 microM). Modafinil 111-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 10757780-5 2000 However, CAR transactivation is increased in the presence of 1,4-bis[2-(3, 5-dichloropyridyloxy)]benzene (TCPOBOP), the most potent known member of the phenobarbital-like class of CYP-inducing agents. 1,4-bis(2-(3,5-dichloropyridyloxy))benzene 61-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 10895987-0 2000 Inhibitory effects of silibinin on cytochrome P-450 enzymes in human liver microsomes. Silybin 22-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 10895987-1 2000 Silibinin, the main constituent of silymarin, a flavonoid drug from silybum marianum used in liver disease, was tested for inhibition of human cytochrome P-450 enzymes. Silybin 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-159 12526496-0 2000 Nitric oxide binding to ferric cytochrome P450: a computational study. Nitric Oxide 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 12526496-1 2000 The interaction between nitric oxide (NO) and the active site of ferric cytochrome P450 was studied by means of density functional theory (DFT), at the generalized gradient approximation level, and of the SAM1 semiempirical method. Nitric Oxide 24-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 10815927-2 2000 The objective of this study was to determine the oxidative metabolites of CPT-11 recovered in human urine samples and to identify cytochrome P450 (CYP) involved in their formation. Irinotecan 74-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 10877005-0 2000 Fluvoxamine but not citalopram increases serum melatonin in healthy subjects-- an indication that cytochrome P450 CYP1A2 and CYP2C19 hydroxylate melatonin. Fluvoxamine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 10877005-0 2000 Fluvoxamine but not citalopram increases serum melatonin in healthy subjects-- an indication that cytochrome P450 CYP1A2 and CYP2C19 hydroxylate melatonin. Melatonin 47-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 10877005-0 2000 Fluvoxamine but not citalopram increases serum melatonin in healthy subjects-- an indication that cytochrome P450 CYP1A2 and CYP2C19 hydroxylate melatonin. Melatonin 145-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 10877007-2 2000 We investigated potential interactions of losartan, irbesartan, valsartan, eprosartan and candesartan with cytochrome P450 (CYP) enzymes in human liver microsomes. Losartan 42-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 10877007-2 2000 We investigated potential interactions of losartan, irbesartan, valsartan, eprosartan and candesartan with cytochrome P450 (CYP) enzymes in human liver microsomes. Valsartan 64-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 10877007-2 2000 We investigated potential interactions of losartan, irbesartan, valsartan, eprosartan and candesartan with cytochrome P450 (CYP) enzymes in human liver microsomes. candesartan 90-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-122 10877007-2 2000 We investigated potential interactions of losartan, irbesartan, valsartan, eprosartan and candesartan with cytochrome P450 (CYP) enzymes in human liver microsomes. candesartan 90-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 10773020-6 2000 The increase in HO activity due to gene transfer also was associated with a parallel decrease (approximately 25%) in cytochrome P-450 (CYP) content and in CYP-dependent arachidonic acid metabolism. Arachidonic Acid 169-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-158 10773020-9 2000 The increase in HO activity after adenoviral-mediated human HO-1 transfer was associated with a decrease in microsomal heme-CYP and CYP activity. Heme 119-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 10797280-2 2000 This study was designed to determine the effects of smoking and 3 major cytochrome P450 (CYP) enzymes, i.e., CYP1A1, CYP1B1 and CYP3A, which metabolize polycyclic aromatic hydrocarbons (PAH) on PAH-DNA adduct formation in the bronchoalveolar macrophages (BAM) of 31 smokers and 16 non-smokers. Polycyclic Aromatic Hydrocarbons 152-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 10797280-2 2000 This study was designed to determine the effects of smoking and 3 major cytochrome P450 (CYP) enzymes, i.e., CYP1A1, CYP1B1 and CYP3A, which metabolize polycyclic aromatic hydrocarbons (PAH) on PAH-DNA adduct formation in the bronchoalveolar macrophages (BAM) of 31 smokers and 16 non-smokers. Polycyclic Aromatic Hydrocarbons 152-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 10797280-2 2000 This study was designed to determine the effects of smoking and 3 major cytochrome P450 (CYP) enzymes, i.e., CYP1A1, CYP1B1 and CYP3A, which metabolize polycyclic aromatic hydrocarbons (PAH) on PAH-DNA adduct formation in the bronchoalveolar macrophages (BAM) of 31 smokers and 16 non-smokers. Polycyclic Aromatic Hydrocarbons 186-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 10797280-2 2000 This study was designed to determine the effects of smoking and 3 major cytochrome P450 (CYP) enzymes, i.e., CYP1A1, CYP1B1 and CYP3A, which metabolize polycyclic aromatic hydrocarbons (PAH) on PAH-DNA adduct formation in the bronchoalveolar macrophages (BAM) of 31 smokers and 16 non-smokers. Polycyclic Aromatic Hydrocarbons 186-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 10797280-2 2000 This study was designed to determine the effects of smoking and 3 major cytochrome P450 (CYP) enzymes, i.e., CYP1A1, CYP1B1 and CYP3A, which metabolize polycyclic aromatic hydrocarbons (PAH) on PAH-DNA adduct formation in the bronchoalveolar macrophages (BAM) of 31 smokers and 16 non-smokers. Polycyclic Aromatic Hydrocarbons 194-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 10797280-2 2000 This study was designed to determine the effects of smoking and 3 major cytochrome P450 (CYP) enzymes, i.e., CYP1A1, CYP1B1 and CYP3A, which metabolize polycyclic aromatic hydrocarbons (PAH) on PAH-DNA adduct formation in the bronchoalveolar macrophages (BAM) of 31 smokers and 16 non-smokers. Polycyclic Aromatic Hydrocarbons 194-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 10833273-9 2000 CYP 4F2 was distinguished from the closely homologous CYP 4F3 (human neutrophil LTB(4) omega-hydroxylase) by its much higher K(m) for LTB(4), inability to omega-hydroxylate lipoxin B(4), and extreme instability. Leukotriene B4 80-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-61 10833273-9 2000 CYP 4F2 was distinguished from the closely homologous CYP 4F3 (human neutrophil LTB(4) omega-hydroxylase) by its much higher K(m) for LTB(4), inability to omega-hydroxylate lipoxin B(4), and extreme instability. Lipoxin B 173-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-61 10833273-9 2000 CYP 4F2 was distinguished from the closely homologous CYP 4F3 (human neutrophil LTB(4) omega-hydroxylase) by its much higher K(m) for LTB(4), inability to omega-hydroxylate lipoxin B(4), and extreme instability. Lipoxin B 173-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-104 10807595-5 2000 The lipoxygenase metabolite 12(S)-hydroxyeicosatetraenoic acid (HETE) and the cytochrome P-450 metabolite 20-HETE both inhibited PCT Na(+)-K(+)-ATPase in a protein kinase C-dependent manner, but the effect was significantly more pronounced in infant PCT. 20-Hete 106-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 10807595-5 2000 The lipoxygenase metabolite 12(S)-hydroxyeicosatetraenoic acid (HETE) and the cytochrome P-450 metabolite 20-HETE both inhibited PCT Na(+)-K(+)-ATPase in a protein kinase C-dependent manner, but the effect was significantly more pronounced in infant PCT. Polychloroterphenyl Compounds 129-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 10757780-5 2000 However, CAR transactivation is increased in the presence of 1,4-bis[2-(3, 5-dichloropyridyloxy)]benzene (TCPOBOP), the most potent known member of the phenobarbital-like class of CYP-inducing agents. 1,4-bis(2-(3,5-dichloropyridyloxy))benzene 106-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 10757780-5 2000 However, CAR transactivation is increased in the presence of 1,4-bis[2-(3, 5-dichloropyridyloxy)]benzene (TCPOBOP), the most potent known member of the phenobarbital-like class of CYP-inducing agents. Phenobarbital 152-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 10729207-1 2000 Expression of human cytochrome P450 (P450) 2B6 in Escherichia coli was achieved following supplementation of the expression medium with chloramphenicol. Chloramphenicol 136-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-46 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. Selegiline 96-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. Selegiline 96-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. Selegiline 96-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 236-239 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. desmethylselegiline 110-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. desmethylselegiline 110-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. Levmetamfetamine 134-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. Levmetamfetamine 134-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. Selegiline 119-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. Selegiline 119-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. desmethylselegiline 183-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. desmethylselegiline 183-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. Levmetamfetamine 207-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 10862503-2 2000 In the current work we have studied the cytochrome P450 (CYP)-catalyzed oxidative metabolism of selegiline to desmethylselegiline and 1-methamphetamine and the effects of selegiline, desmethylselegiline and 1-methamphetamine on hepatic CYP enzymes in human liver microsomes in vitro. Levmetamfetamine 207-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 10862503-8 2000 In studies with CYP-specific model activities, both selegiline and desmethylselegiline inhibited the CYP2C19-mediated S-mephenytoin 4"-hydroxylation with average IC50 values of 21 microM and 26 microM, respectively. Selegiline 52-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 10862503-8 2000 In studies with CYP-specific model activities, both selegiline and desmethylselegiline inhibited the CYP2C19-mediated S-mephenytoin 4"-hydroxylation with average IC50 values of 21 microM and 26 microM, respectively. desmethylselegiline 67-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 10862503-11 2000 Inhibitory potencies of selegiline, desmethylselegiline and 1-methamphetamine towards other CYP-model activities were much lower. Selegiline 24-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 10862503-11 2000 Inhibitory potencies of selegiline, desmethylselegiline and 1-methamphetamine towards other CYP-model activities were much lower. desmethylselegiline 36-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 10862503-11 2000 Inhibitory potencies of selegiline, desmethylselegiline and 1-methamphetamine towards other CYP-model activities were much lower. Levmetamfetamine 60-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-95 11055180-4 2000 These are: microsomal ethanol oxidation system (MEOS) connected with cytochrome P-450 and peroxisome catalase system. Ethanol 22-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 11239797-1 2000 Studies designed to elucidate the identity of the potent vasodilator autacoid endothelium-derived hyperpolarizing factor (EDHF) in the coronary vascular bed have highlighted a role for vascular cytochrome P450 (CYP) enzymes in cardiovascular homeostasis. hyperpolarizing factor 98-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-209 11239797-1 2000 Studies designed to elucidate the identity of the potent vasodilator autacoid endothelium-derived hyperpolarizing factor (EDHF) in the coronary vascular bed have highlighted a role for vascular cytochrome P450 (CYP) enzymes in cardiovascular homeostasis. hyperpolarizing factor 98-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-214 11239797-1 2000 Studies designed to elucidate the identity of the potent vasodilator autacoid endothelium-derived hyperpolarizing factor (EDHF) in the coronary vascular bed have highlighted a role for vascular cytochrome P450 (CYP) enzymes in cardiovascular homeostasis. edhf 122-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-209 11239797-1 2000 Studies designed to elucidate the identity of the potent vasodilator autacoid endothelium-derived hyperpolarizing factor (EDHF) in the coronary vascular bed have highlighted a role for vascular cytochrome P450 (CYP) enzymes in cardiovascular homeostasis. edhf 122-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-214 11239797-2 2000 Not only is there strong evidence suggesting that the putative coronary EDHF synthase is an endothelially expressed CYP epoxygenase, but CYP products such as epoxyeicosatrienoic acids and reactive oxygen species have been implicated in the regulation of intracellular signalling cascades and vascular cell proliferation. epoxyeicosatrienoic acids 158-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 10826653-1 2000 Our previous results indicated that cytochrome P450 destruction by benzene metabolites was caused mainly by benzoquinone (Soucek et al., Biochem. Benzene 67-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 10826653-1 2000 Our previous results indicated that cytochrome P450 destruction by benzene metabolites was caused mainly by benzoquinone (Soucek et al., Biochem. quinone 108-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 10819168-0 2000 The use of "electronic nose" sensor responses to predict the inhibition activity of alcohols on the cytochrome P-450 catalyzed p-hydroxylation of aniline. Alcohols 84-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 10759690-0 2000 In vitro inhibition of the cytochrome P450 (CYP450) system by the antiplatelet drug ticlopidine: potent effect on CYP2C19 and CYP2D6. Ticlopidine 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 10759690-0 2000 In vitro inhibition of the cytochrome P450 (CYP450) system by the antiplatelet drug ticlopidine: potent effect on CYP2C19 and CYP2D6. Ticlopidine 84-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-50 10819168-0 2000 The use of "electronic nose" sensor responses to predict the inhibition activity of alcohols on the cytochrome P-450 catalyzed p-hydroxylation of aniline. aniline 146-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 10819168-1 2000 A quantitative structure activity relationship (QSAR) has been formulated to describe the inhibitory action of a series of alcohols on the cytochrome P-450 catalyzed p-hydroxylation of aniline. Alcohols 123-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 10819168-1 2000 A quantitative structure activity relationship (QSAR) has been formulated to describe the inhibitory action of a series of alcohols on the cytochrome P-450 catalyzed p-hydroxylation of aniline. aniline 185-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 10819168-3 2000 If the various electronic nose sensor response patterns for the family of test alcohols reflect differences in the chemical properties that are involved in the cytochrome P-450 inhibition process, it ought to be possible to correlate the differences in the electronic nose signals of these analytes with the differences in the cytochrome P-450 inhibition by these species. Alcohols 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 10819168-3 2000 If the various electronic nose sensor response patterns for the family of test alcohols reflect differences in the chemical properties that are involved in the cytochrome P-450 inhibition process, it ought to be possible to correlate the differences in the electronic nose signals of these analytes with the differences in the cytochrome P-450 inhibition by these species. Alcohols 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 327-343 10681373-0 2000 Human cytochrome P-450 metabolism of retinals to retinoic acids. Tretinoin 49-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 10725303-2 2000 We previously showed that liver volume normalized to body weight was inversely related to age, but that the systemic clearance of a nonspecific cytochrome P450 (CYP) substrate (antipyrine) was higher in young children compared with adults even when normalized per liver volume. Antipyrine 177-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-159 10725303-2 2000 We previously showed that liver volume normalized to body weight was inversely related to age, but that the systemic clearance of a nonspecific cytochrome P450 (CYP) substrate (antipyrine) was higher in young children compared with adults even when normalized per liver volume. Antipyrine 177-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-164 10725304-2 2000 The human cytochrome P450 (CYP) involved in bupivacaine degradation into pipecolylxylidine (PPX), its major metabolite, has, to our knowledge, never been described. Bupivacaine 44-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 10725304-2 2000 The human cytochrome P450 (CYP) involved in bupivacaine degradation into pipecolylxylidine (PPX), its major metabolite, has, to our knowledge, never been described. Bupivacaine 44-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 10725304-2 2000 The human cytochrome P450 (CYP) involved in bupivacaine degradation into pipecolylxylidine (PPX), its major metabolite, has, to our knowledge, never been described. pipecolylxylidine 73-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 10725304-2 2000 The human cytochrome P450 (CYP) involved in bupivacaine degradation into pipecolylxylidine (PPX), its major metabolite, has, to our knowledge, never been described. pipecolylxylidine 73-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 10725304-2 2000 The human cytochrome P450 (CYP) involved in bupivacaine degradation into pipecolylxylidine (PPX), its major metabolite, has, to our knowledge, never been described. ppx 92-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 10725304-2 2000 The human cytochrome P450 (CYP) involved in bupivacaine degradation into pipecolylxylidine (PPX), its major metabolite, has, to our knowledge, never been described. ppx 92-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 10725304-5 2000 Bupivacaine incubations were then performed with specific CYP substrates and inhibitors. Bupivacaine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 10725309-0 2000 Studies on the cytochrome P450 (CYP)-mediated metabolic properties of miocamycin: evaluation of the possibility of a metabolic intermediate complex formation with CYP, and identification of the human CYP isoforms. Miocamycin 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-30 10725309-0 2000 Studies on the cytochrome P450 (CYP)-mediated metabolic properties of miocamycin: evaluation of the possibility of a metabolic intermediate complex formation with CYP, and identification of the human CYP isoforms. Miocamycin 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 10725309-0 2000 Studies on the cytochrome P450 (CYP)-mediated metabolic properties of miocamycin: evaluation of the possibility of a metabolic intermediate complex formation with CYP, and identification of the human CYP isoforms. Miocamycin 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-166 10725309-0 2000 Studies on the cytochrome P450 (CYP)-mediated metabolic properties of miocamycin: evaluation of the possibility of a metabolic intermediate complex formation with CYP, and identification of the human CYP isoforms. Miocamycin 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-166 10725309-1 2000 Some macrolide antibiotics cause clinical drug interactions, resulting in altered metabolism of concomitantly administered drugs, via the formation of a metabolic intermediate (MI) complex with cytochrome P450 (CYP), or competitive inhibition of CYP. macrolide antibiotics 5-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-209 10725309-1 2000 Some macrolide antibiotics cause clinical drug interactions, resulting in altered metabolism of concomitantly administered drugs, via the formation of a metabolic intermediate (MI) complex with cytochrome P450 (CYP), or competitive inhibition of CYP. macrolide antibiotics 5-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-214 10725309-1 2000 Some macrolide antibiotics cause clinical drug interactions, resulting in altered metabolism of concomitantly administered drugs, via the formation of a metabolic intermediate (MI) complex with cytochrome P450 (CYP), or competitive inhibition of CYP. macrolide antibiotics 5-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-249 10681376-10 2000 Based on responses to selective chemical inhibitors, specific activities, and levels present in adult human hepatic tissues, CYP1A2 and CYP3A4 strongly appeared to be the major CYP enzymes catalyzing hepatic oxidative conversion of t-ROH to t-RAL in the adult human liver. t-roh 232-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 10681382-1 2000 Tolbutamide is a sulfonylurea-type oral hypoglycemic agent whose action is terminated by hydroxylation of the tolylsulfonyl methyl moiety catalyzed by cytochrome P-450 (CYP) enzymes of the human CYP2C subfamily. Tolbutamide 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 10681382-1 2000 Tolbutamide is a sulfonylurea-type oral hypoglycemic agent whose action is terminated by hydroxylation of the tolylsulfonyl methyl moiety catalyzed by cytochrome P-450 (CYP) enzymes of the human CYP2C subfamily. Tolbutamide 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 10681382-1 2000 Tolbutamide is a sulfonylurea-type oral hypoglycemic agent whose action is terminated by hydroxylation of the tolylsulfonyl methyl moiety catalyzed by cytochrome P-450 (CYP) enzymes of the human CYP2C subfamily. Sulfonylurea Compounds 17-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 10681382-1 2000 Tolbutamide is a sulfonylurea-type oral hypoglycemic agent whose action is terminated by hydroxylation of the tolylsulfonyl methyl moiety catalyzed by cytochrome P-450 (CYP) enzymes of the human CYP2C subfamily. Sulfonylurea Compounds 17-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 10780971-3 2000 The aims of this study were to investigate the metabolism of cisapride in human liver microsomes and to determine which cytochrome P-450 (CYP) isoenzyme(s) are involved in cisapride biotransformation. Cisapride 172-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 10780971-3 2000 The aims of this study were to investigate the metabolism of cisapride in human liver microsomes and to determine which cytochrome P-450 (CYP) isoenzyme(s) are involved in cisapride biotransformation. Cisapride 172-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 10764440-2 2000 Particular emphasis is placed on describing the newer information on the cytochrome P450 (CYP) system and the interactions of opioids, antidepressants, and the antitussive, dextromethorphan. Dextromethorphan 173-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-93 10681382-1 2000 Tolbutamide is a sulfonylurea-type oral hypoglycemic agent whose action is terminated by hydroxylation of the tolylsulfonyl methyl moiety catalyzed by cytochrome P-450 (CYP) enzymes of the human CYP2C subfamily. tolylsulfonyl 110-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 10681376-2 2000 The purpose of this study was to investigate the role of human cytochrome P-450 (CYP)-dependent monooxygenation in the conversion of t-ROH to t-RAL. t-roh 133-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 10681382-1 2000 Tolbutamide is a sulfonylurea-type oral hypoglycemic agent whose action is terminated by hydroxylation of the tolylsulfonyl methyl moiety catalyzed by cytochrome P-450 (CYP) enzymes of the human CYP2C subfamily. tolylsulfonyl 110-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 10681376-2 2000 The purpose of this study was to investigate the role of human cytochrome P-450 (CYP)-dependent monooxygenation in the conversion of t-ROH to t-RAL. t-roh 133-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-84 10640505-0 2000 Novel metabolic pathway of estrone and 17beta-estradiol catalyzed by cytochrome P-450. Estrone 27-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 10706031-4 2000 These findings are not inconsistent with the hypothesis derived from epidemiological work and animal studies that organochlorine insecticides produce a direct toxic action on the dopaminergic tracts of the substantia nigra and may contribute to the development of PD in those rendered susceptible by virtue of cytochrome P-450 polymorphism, excessive exposure, or other factors. Hydrocarbons, Chlorinated 114-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 310-326 10746939-4 2000 Our laboratory is using the human H295R adrenocortical carcinoma cell line to examine chemicals for their potential to interfere with the activity and/or expression of several key cytochrome P450 (CYP) enzymes involved in the biosynthesis of steroid hormones. Steroids 242-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-195 10746939-4 2000 Our laboratory is using the human H295R adrenocortical carcinoma cell line to examine chemicals for their potential to interfere with the activity and/or expression of several key cytochrome P450 (CYP) enzymes involved in the biosynthesis of steroid hormones. Steroids 242-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-200 10640505-0 2000 Novel metabolic pathway of estrone and 17beta-estradiol catalyzed by cytochrome P-450. Estradiol 39-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 10640505-1 2000 We have already reported that the quinol formation from some para-alkylphenols, which is a novel metabolic pathway catalyzed by cytochrome P-450, occurs in a rat liver microsomal system (). Hydroquinones 34-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 10640505-1 2000 We have already reported that the quinol formation from some para-alkylphenols, which is a novel metabolic pathway catalyzed by cytochrome P-450, occurs in a rat liver microsomal system (). para-alkylphenols 61-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-144 10640505-2 2000 In the present study, we investigated whether estrone and 17beta-estadiol, each of which contains a p-alkylphenol moiety, are also oxidized into the corresponding quinols by cytochrome P-450. Estrone 46-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-190 10640505-2 2000 In the present study, we investigated whether estrone and 17beta-estadiol, each of which contains a p-alkylphenol moiety, are also oxidized into the corresponding quinols by cytochrome P-450. 17beta-estadiol 58-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-190 10640505-2 2000 In the present study, we investigated whether estrone and 17beta-estadiol, each of which contains a p-alkylphenol moiety, are also oxidized into the corresponding quinols by cytochrome P-450. Hydroquinones 163-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-190 11390867-2 2000 In the coronary circulation, this endothelium-derived hyperpolarizing factor appears to be a cytochrome P(450)-derived arachidonic acid epoxide, the release of which may play a crucial role in the maintenance of coronary blood flow in arteriosclerosis. arachidonic acid epoxide 119-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-111 10805063-15 2000 The inhibition of human CYP isoforms by 1,4-dihydropyridine calcium antagonists except nicardipine might be clinically insignificant. 1,4-dihydropyridine calcium 40-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 10805064-1 2000 OBJECTIVE: Halothane undergoes both oxidative and reductive metabolism by cytochrome P450 (CYP), respectively causing rare immune-mediated hepatic necrosis and common, mild subclinical hepatic toxicity. Halothane 11-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 10805064-1 2000 OBJECTIVE: Halothane undergoes both oxidative and reductive metabolism by cytochrome P450 (CYP), respectively causing rare immune-mediated hepatic necrosis and common, mild subclinical hepatic toxicity. Halothane 11-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-94 10805064-15 2000 CYP2A6-mediated, like CYP2E1-mediated human halothane oxidation, can be inhibited in vivo by mechanism-based CYP inhibitors. Halothane 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 10698112-0 2000 Cytochrome P450 catalyzed nitric oxide synthesis: a theoretical study. Nitric Oxide 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10698112-1 2000 Similar to nitric oxide synthase (NOS) cytochrome P450 isoforms (e.g. 3A and 4E) can produce nitric oxide from arginine. Nitric Oxide 11-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 10698112-1 2000 Similar to nitric oxide synthase (NOS) cytochrome P450 isoforms (e.g. 3A and 4E) can produce nitric oxide from arginine. Arginine 111-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 10698112-7 2000 The lowest energy conformation of the cytochrome P450 3A4-substrate complex was compared to the high resolution X-ray structure of the iNOS-arginine complex. Arginine 140-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 10681399-2 2000 The demonstration of in vivo arachidonic acid epoxidation and omega-hydroxylation established the cytochrome P450 epoxygenase and omega/omega-1 hydroxylase as formal metabolic pathways and as members of the arachidonate metabolic cascade. Arachidonic Acid 29-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 10681399-2 2000 The demonstration of in vivo arachidonic acid epoxidation and omega-hydroxylation established the cytochrome P450 epoxygenase and omega/omega-1 hydroxylase as formal metabolic pathways and as members of the arachidonate metabolic cascade. Arachidonic Acid 207-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-113 10681399-3 2000 The characterization of the potent biological activities associated with several of the cytochrome P450-derived eicosanoids suggested new and important functional roles for these enzymes in cellular, organ, and body physiology, including the control of vascular reactivity and systemic blood pressures. Eicosanoids 112-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 10681399-4 2000 Past and current advances in cytochrome P450 biochemistry and molecular biology facilitate the characterization of cytochrome P450 isoforms responsible for tissue/organ specific arachidonic acid epoxidation and omega/omega-1 hydroxylation, and thus, the analysis of cDNA and/or gene specific functional phenotypes. Arachidonic Acid 178-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 10681399-4 2000 Past and current advances in cytochrome P450 biochemistry and molecular biology facilitate the characterization of cytochrome P450 isoforms responsible for tissue/organ specific arachidonic acid epoxidation and omega/omega-1 hydroxylation, and thus, the analysis of cDNA and/or gene specific functional phenotypes. Arachidonic Acid 178-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-130 10805063-0 2000 Inhibition of human cytochrome P450 enzymes by 1,4-dihydropyridine calcium antagonists: prediction of in vivo drug-drug interactions. 1,4-dihydropyridine calcium 47-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 10805063-2 2000 It is now commonly agreed that the oxidation of 1,4-dihydropyridine into pyridine, which is one of the main metabolic pathways, is catalysed by the cytochrome P450 (CYP) 3A4 isoform. 1,4-dihydropyridine 48-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-163 10805063-2 2000 It is now commonly agreed that the oxidation of 1,4-dihydropyridine into pyridine, which is one of the main metabolic pathways, is catalysed by the cytochrome P450 (CYP) 3A4 isoform. 1,4-dihydropyridine 48-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-168 10805063-2 2000 It is now commonly agreed that the oxidation of 1,4-dihydropyridine into pyridine, which is one of the main metabolic pathways, is catalysed by the cytochrome P450 (CYP) 3A4 isoform. pyridine 59-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-163 10805063-2 2000 It is now commonly agreed that the oxidation of 1,4-dihydropyridine into pyridine, which is one of the main metabolic pathways, is catalysed by the cytochrome P450 (CYP) 3A4 isoform. pyridine 59-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-168 10805063-3 2000 In the present study, the inhibitory effects of 13 kinds of 1,4-dihydropyridine calcium antagonists clinically used in Japan on human CYP-isoform-dependent reactions were investigated to predict the drug interactions using microsomes from human B-lymphoblast cells expressing CYP. 1,4-dihydropyridine calcium 60-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-137 10805063-12 2000 Among the human CYP isoforms investigated, the inhibitory effects of 1,4-dihydropyridine calcium antagonists were potent on human CYP1A2, CYP2B6, CYP2C9, CYP2C19 and CYP2D6 as well as CYP3A4. 1,4-dihydropyridine calcium 69-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 10653206-0 2000 Fluvoxamine-Clozapine drug interaction: inhibition in vitro of five cytochrome P450 isoforms involved in clozapine metabolism. Fluvoxamine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 10653206-0 2000 Fluvoxamine-Clozapine drug interaction: inhibition in vitro of five cytochrome P450 isoforms involved in clozapine metabolism. Clozapine 12-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 10653206-0 2000 Fluvoxamine-Clozapine drug interaction: inhibition in vitro of five cytochrome P450 isoforms involved in clozapine metabolism. Clozapine 105-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 10653206-5 2000 Fluvoxamine also inhibited in a concentration-dependent manner the activity of all five cytochrome P450 (CYP) isoforms previously determined to be capable of catalyzing the demethylation of clozapine. Fluvoxamine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 10653206-5 2000 Fluvoxamine also inhibited in a concentration-dependent manner the activity of all five cytochrome P450 (CYP) isoforms previously determined to be capable of catalyzing the demethylation of clozapine. Fluvoxamine 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-108 10653206-5 2000 Fluvoxamine also inhibited in a concentration-dependent manner the activity of all five cytochrome P450 (CYP) isoforms previously determined to be capable of catalyzing the demethylation of clozapine. Clozapine 190-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 10653206-5 2000 Fluvoxamine also inhibited in a concentration-dependent manner the activity of all five cytochrome P450 (CYP) isoforms previously determined to be capable of catalyzing the demethylation of clozapine. Clozapine 190-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-108 10709852-5 2000 Down-regulation of CYP 2C protein by transfection of porcine coronary arteries with anti-sense oligonucleotides decreased EDHF-mediated vascular responses while EDHF-mediated hyperpolarisation and relaxation were potentiated by the CYP-inducing compound beta-naphthoflavone. edhf 122-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 10696794-2 2000 Oxidative metabolism of Tri by cytochrome P-450 to form chloral hydrate (CH) was only detectable in kidney microsomes from one patient out of four tested and was not detected in hPT cells. Chloral Hydrate 56-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 10709852-0 2000 EDHF: a cytochrome P450 metabolite in coronary arteries. edhf 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-23 10709852-3 2000 In coronary arteries EDHF has been pharmacologically characterized as a cytochrome P450 (CYP)-derived metabolite of arachidonic acid. Arachidonic Acid 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 10709852-5 2000 Down-regulation of CYP 2C protein by transfection of porcine coronary arteries with anti-sense oligonucleotides decreased EDHF-mediated vascular responses while EDHF-mediated hyperpolarisation and relaxation were potentiated by the CYP-inducing compound beta-naphthoflavone. edhf 161-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 10709852-3 2000 In coronary arteries EDHF has been pharmacologically characterized as a cytochrome P450 (CYP)-derived metabolite of arachidonic acid. Arachidonic Acid 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 10709852-5 2000 Down-regulation of CYP 2C protein by transfection of porcine coronary arteries with anti-sense oligonucleotides decreased EDHF-mediated vascular responses while EDHF-mediated hyperpolarisation and relaxation were potentiated by the CYP-inducing compound beta-naphthoflavone. Oligonucleotides 95-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 10709852-5 2000 Down-regulation of CYP 2C protein by transfection of porcine coronary arteries with anti-sense oligonucleotides decreased EDHF-mediated vascular responses while EDHF-mediated hyperpolarisation and relaxation were potentiated by the CYP-inducing compound beta-naphthoflavone. beta-Naphthoflavone 254-273 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 10709852-6 2000 Thus, CYP 2C appears to play a crucial role in the generation of EDHF-mediated responses in porcine coronary arteries. edhf 65-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 11268336-6 2000 Most recently, evidence has accumulated that cytochrome P-450 (P-450), part of the alternative pathway for arachidonic acid metabolism, plays an important role in reduction of fever and inflammation. Arachidonic Acid 107-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 10771452-0 2000 Identification of the major human liver cytochrome P450 isoform(s) responsible for the formation of the primary metabolites of ziprasidone and prediction of possible drug interactions. ziprasidone 127-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 10911933-3 2000 Cytochrome P450 drug metabolizing enzymes (CYPs), can activate (e.g. codeine to morphine) or deactivate (e.g. nicotine to cotinine) drugs of abuse. Morphine 80-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10911933-3 2000 Cytochrome P450 drug metabolizing enzymes (CYPs), can activate (e.g. codeine to morphine) or deactivate (e.g. nicotine to cotinine) drugs of abuse. Nicotine 110-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10911933-3 2000 Cytochrome P450 drug metabolizing enzymes (CYPs), can activate (e.g. codeine to morphine) or deactivate (e.g. nicotine to cotinine) drugs of abuse. Cotinine 122-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10771452-13 2000 CONCLUSIONS: Ziprasidone is predominantly metabolized by CYP3A4 in human liver microsomes and is not expected to mediate drug interactions with coadministered CYP substrates, at clinically effective doses. ziprasidone 13-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 10911933-3 2000 Cytochrome P450 drug metabolizing enzymes (CYPs), can activate (e.g. codeine to morphine) or deactivate (e.g. nicotine to cotinine) drugs of abuse. Codeine 69-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10771452-1 2000 AIMS: To identify the cytochrome P450 (CYP) isoform(s) responsible for the formation of the primary metabolite of ziprasidone (ziprasidone sulphoxide), to determine the kinetics of its formation and to predict possible drug interactions by investigating CYP isoform inhibition in an in vitro study. ziprasidone 114-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 10771452-1 2000 AIMS: To identify the cytochrome P450 (CYP) isoform(s) responsible for the formation of the primary metabolite of ziprasidone (ziprasidone sulphoxide), to determine the kinetics of its formation and to predict possible drug interactions by investigating CYP isoform inhibition in an in vitro study. ziprasidone 114-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 10771452-1 2000 AIMS: To identify the cytochrome P450 (CYP) isoform(s) responsible for the formation of the primary metabolite of ziprasidone (ziprasidone sulphoxide), to determine the kinetics of its formation and to predict possible drug interactions by investigating CYP isoform inhibition in an in vitro study. ziprasidone 114-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 254-257 10771452-1 2000 AIMS: To identify the cytochrome P450 (CYP) isoform(s) responsible for the formation of the primary metabolite of ziprasidone (ziprasidone sulphoxide), to determine the kinetics of its formation and to predict possible drug interactions by investigating CYP isoform inhibition in an in vitro study. Ziprasidone Sulfoxide 127-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-37 10771452-1 2000 AIMS: To identify the cytochrome P450 (CYP) isoform(s) responsible for the formation of the primary metabolite of ziprasidone (ziprasidone sulphoxide), to determine the kinetics of its formation and to predict possible drug interactions by investigating CYP isoform inhibition in an in vitro study. Ziprasidone Sulfoxide 127-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-42 10771452-1 2000 AIMS: To identify the cytochrome P450 (CYP) isoform(s) responsible for the formation of the primary metabolite of ziprasidone (ziprasidone sulphoxide), to determine the kinetics of its formation and to predict possible drug interactions by investigating CYP isoform inhibition in an in vitro study. Ziprasidone Sulfoxide 127-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 254-257 10771452-7 2000 RESULTS: Three CYP-mediated metabolites - ziprasidone sulphoxide, ziprasidone sulphone and oxindole acetic acid - were identified. Ziprasidone Sulfoxide 42-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 10771452-7 2000 RESULTS: Three CYP-mediated metabolites - ziprasidone sulphoxide, ziprasidone sulphone and oxindole acetic acid - were identified. UNII-6BZX6ST0LP 66-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 11059564-0 2000 Inhibition of the cytochrome P-450 modulates all-trans-retinoic acid-induced differentiation and apoptosis of HL-60 cells. 2-octenal 49-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 10771452-7 2000 RESULTS: Three CYP-mediated metabolites - ziprasidone sulphoxide, ziprasidone sulphone and oxindole acetic acid - were identified. oxindole acetic acid 91-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 11059564-0 2000 Inhibition of the cytochrome P-450 modulates all-trans-retinoic acid-induced differentiation and apoptosis of HL-60 cells. Tretinoin 55-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 11059564-1 2000 We studied the effects of inhibition of cytochrome P-450 by proadifen (SKF525A) on the processes induced in myeloid leukemia HL-60 cells by all-trans-retinoic acid (ATRA). Proadifen 71-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 11122720-1 2000 The cytochrome P450 (CYP) is a group of enzymes that oxidatively modify drugs to a more water-soluble form for renal excretion. Water 88-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 10747392-8 2000 For anisol and thioanisol, analogies and differences between oxidation reactions catalyzed by the enzyme cytochrome P-450 in the condensed phase and those observed for the gas-phase model FeO+ are discussed. methylphenylsulfide 15-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 11122720-1 2000 The cytochrome P450 (CYP) is a group of enzymes that oxidatively modify drugs to a more water-soluble form for renal excretion. Water 88-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 11122720-2 2000 Nearly 50% of all clinically used medications and endogenous steroids are metabolized by the CYP enzyme 3A4, which explains why many of the important potential drug interactions involved this enzyme. Steroids 61-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 11122720-3 2000 Despite an excellent safety record, CYP 3A4 statins (lovastatin, simvastatin, atorvastatin) taken concomitantly with a potent CYP 3A4 inhibitor may increase the risk for adverse events (myopathy, rhabdomyolysis). Lovastatin 53-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 11098412-4 2000 Based on these parameters, selecting an antidepressant medication, such as venlafaxine, that has a low potential for drug interactions at the Cytochrome P450 (CYP) enzyme system, and is easy to monitor and dose, facilitate successful treatment of patients. Venlafaxine Hydrochloride 75-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 11098412-4 2000 Based on these parameters, selecting an antidepressant medication, such as venlafaxine, that has a low potential for drug interactions at the Cytochrome P450 (CYP) enzyme system, and is easy to monitor and dose, facilitate successful treatment of patients. Venlafaxine Hydrochloride 75-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-162 11122720-3 2000 Despite an excellent safety record, CYP 3A4 statins (lovastatin, simvastatin, atorvastatin) taken concomitantly with a potent CYP 3A4 inhibitor may increase the risk for adverse events (myopathy, rhabdomyolysis). Simvastatin 65-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 11122720-3 2000 Despite an excellent safety record, CYP 3A4 statins (lovastatin, simvastatin, atorvastatin) taken concomitantly with a potent CYP 3A4 inhibitor may increase the risk for adverse events (myopathy, rhabdomyolysis). Atorvastatin 78-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 11122720-3 2000 Despite an excellent safety record, CYP 3A4 statins (lovastatin, simvastatin, atorvastatin) taken concomitantly with a potent CYP 3A4 inhibitor may increase the risk for adverse events (myopathy, rhabdomyolysis). Atorvastatin 78-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 11122720-4 2000 This article describes the clinical significance of CYP metabolism as the pathways relate to the use of statins, including brief discussions on statins, fibrates, cyclosporine, and calcium channel blockers. Cyclosporine 163-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-55 10611138-10 2000 These data suggest that acetone and acetonitrile stimulate NADPH-mediated tolbutamide hydroxylation via the CYP reductase and not by modifying the affinity of tolbutamide for the CYP2C9 enzyme. Acetone 24-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 10601869-1 2000 An 1H-NMR study of ferric cytochrome P450cam in different paramagnetic states was performed. Hydrogen 3-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 10611138-10 2000 These data suggest that acetone and acetonitrile stimulate NADPH-mediated tolbutamide hydroxylation via the CYP reductase and not by modifying the affinity of tolbutamide for the CYP2C9 enzyme. acetonitrile 36-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 10611138-10 2000 These data suggest that acetone and acetonitrile stimulate NADPH-mediated tolbutamide hydroxylation via the CYP reductase and not by modifying the affinity of tolbutamide for the CYP2C9 enzyme. NADP 59-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 10611138-10 2000 These data suggest that acetone and acetonitrile stimulate NADPH-mediated tolbutamide hydroxylation via the CYP reductase and not by modifying the affinity of tolbutamide for the CYP2C9 enzyme. Tolbutamide 74-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-111 10611146-0 2000 Effect of the adrenal 11-beta-hydroxylase inhibitor metyrapone on human hepatic cytochrome P-450 expression: induction of cytochrome P-450 3A4. Metyrapone 52-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 10712743-2 2000 GSTP1 and GSTM1 are mainly involved in detoxification reactions of PAH carcinogenic intermediates produced by cytochrome P450 (CYP). p-Aminohippuric Acid 67-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 10712743-2 2000 GSTP1 and GSTM1 are mainly involved in detoxification reactions of PAH carcinogenic intermediates produced by cytochrome P450 (CYP). p-Aminohippuric Acid 67-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 10601869-0 2000 Assignment of heme methyl 1H-NMR resonances of high-spin and low-spin ferric complexes of cytochrome p450cam using one-dimensional and two-dimensional paramagnetic signals enhancement (PASE) magnetization transfer experiments. Heme 14-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 10601869-0 2000 Assignment of heme methyl 1H-NMR resonances of high-spin and low-spin ferric complexes of cytochrome p450cam using one-dimensional and two-dimensional paramagnetic signals enhancement (PASE) magnetization transfer experiments. Hydrogen 26-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 10601869-2 2000 Assignment of three heme methyl resonances of the isocyanide adduct of cytochrome P450 in the ferric low-spin state was recently performed using electron exchange in the presence of putidaredoxin [Mouro, C., Bondon, A., Jung, C., Hui Bon Hoa, G., De Certaines, J.D., Spencer, R.G.S. Heme 20-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 10601869-0 2000 Assignment of heme methyl 1H-NMR resonances of high-spin and low-spin ferric complexes of cytochrome p450cam using one-dimensional and two-dimensional paramagnetic signals enhancement (PASE) magnetization transfer experiments. Ferric enterobactin ion 70-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 10601869-2 2000 Assignment of three heme methyl resonances of the isocyanide adduct of cytochrome P450 in the ferric low-spin state was recently performed using electron exchange in the presence of putidaredoxin [Mouro, C., Bondon, A., Jung, C., Hui Bon Hoa, G., De Certaines, J.D., Spencer, R.G.S. Cyanides 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 10601869-2 2000 Assignment of three heme methyl resonances of the isocyanide adduct of cytochrome P450 in the ferric low-spin state was recently performed using electron exchange in the presence of putidaredoxin [Mouro, C., Bondon, A., Jung, C., Hui Bon Hoa, G., De Certaines, J.D., Spencer, R.G.S. Ferric enterobactin ion 94-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 10601869-5 2000 In this study, heme methyl protons of cytochrome P450 in the native high-spin and low-spin states were assigned through one-dimensional and two-dimensional magnetization transfer spectroscopy using the paramagnetic signals enhancement (PASE) method. Heme 15-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 10690614-2 2000 BACKGROUND/AIMS: Liver function can be evaluated using 13C breath tests that explore liver Cytochrome P450 activity. 13c 55-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 10798223-1 2000 The well-documented reduction of cancer risk by high dietary cruciferous vegetable intake may in part be caused by modulation of cytochrome P-450 (CYP) expression and activity by indoles. Indoles 179-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 10711629-0 2000 Metabolism of thalidomide in human microsomes, cloned human cytochrome P-450 isozymes, and Hansen"s disease patients. Thalidomide 14-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 10798223-1 2000 The well-documented reduction of cancer risk by high dietary cruciferous vegetable intake may in part be caused by modulation of cytochrome P-450 (CYP) expression and activity by indoles. Indoles 179-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 10616189-0 1999 Metabolism of N-nitrosobenzylmethylamine by human cytochrome P-450 enzymes. nitrosobenzylmethylamine 14-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 11013424-3 2000 A highly efficient direct injection on-line guard cartridge extraction/tandem mass spectrometry (DI-GCE/MS/MS) method has been validated for high-throughput evaluation of cytochrome P450 (CYP) 3A4, 2D6 and 2E1 inhibition potential via cassette dosing of midazolam, dextromethorphan and chlorzoxazone using human hepatic microsomes and 96-well microtiter plates. Midazolam 254-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-186 11013424-3 2000 A highly efficient direct injection on-line guard cartridge extraction/tandem mass spectrometry (DI-GCE/MS/MS) method has been validated for high-throughput evaluation of cytochrome P450 (CYP) 3A4, 2D6 and 2E1 inhibition potential via cassette dosing of midazolam, dextromethorphan and chlorzoxazone using human hepatic microsomes and 96-well microtiter plates. Midazolam 254-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-191 11013424-3 2000 A highly efficient direct injection on-line guard cartridge extraction/tandem mass spectrometry (DI-GCE/MS/MS) method has been validated for high-throughput evaluation of cytochrome P450 (CYP) 3A4, 2D6 and 2E1 inhibition potential via cassette dosing of midazolam, dextromethorphan and chlorzoxazone using human hepatic microsomes and 96-well microtiter plates. Dextromethorphan 265-281 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-186 11013424-3 2000 A highly efficient direct injection on-line guard cartridge extraction/tandem mass spectrometry (DI-GCE/MS/MS) method has been validated for high-throughput evaluation of cytochrome P450 (CYP) 3A4, 2D6 and 2E1 inhibition potential via cassette dosing of midazolam, dextromethorphan and chlorzoxazone using human hepatic microsomes and 96-well microtiter plates. Dextromethorphan 265-281 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-191 11013424-3 2000 A highly efficient direct injection on-line guard cartridge extraction/tandem mass spectrometry (DI-GCE/MS/MS) method has been validated for high-throughput evaluation of cytochrome P450 (CYP) 3A4, 2D6 and 2E1 inhibition potential via cassette dosing of midazolam, dextromethorphan and chlorzoxazone using human hepatic microsomes and 96-well microtiter plates. Chlorzoxazone 286-299 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-186 11013424-3 2000 A highly efficient direct injection on-line guard cartridge extraction/tandem mass spectrometry (DI-GCE/MS/MS) method has been validated for high-throughput evaluation of cytochrome P450 (CYP) 3A4, 2D6 and 2E1 inhibition potential via cassette dosing of midazolam, dextromethorphan and chlorzoxazone using human hepatic microsomes and 96-well microtiter plates. Chlorzoxazone 286-299 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-191 10628896-2 1999 The enzymes involved in the biotransformation of haloperidol include cytochrome P450 (CYP), carbonyl reductase and uridine diphosphoglucose glucuronosyltransferase. Haloperidol 49-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 10602224-0 1999 On the "Rebound" Mechanism of Alkane Hydroxylation by Cytochrome P450: Electronic Structure of the Intermediate and the Electron Transfer Character in the Rebound Step. Alkanes 30-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 10602224-1 1999 Two electromeric forms, a and b (a is the ground state in a solvent) exist for the hydroxo-iron complex 1, an intermediate in the rebound mechanism of alkane hydroxylation by cytochrome P450. hydroxo-iron 83-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-190 10602224-1 1999 Two electromeric forms, a and b (a is the ground state in a solvent) exist for the hydroxo-iron complex 1, an intermediate in the rebound mechanism of alkane hydroxylation by cytochrome P450. Alkanes 151-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 175-190 10628896-2 1999 The enzymes involved in the biotransformation of haloperidol include cytochrome P450 (CYP), carbonyl reductase and uridine diphosphoglucose glucuronosyltransferase. Haloperidol 49-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 10628896-3 1999 The greatest proportion of the intrinsic hepatic clearance of haloperidol is by glucuronidation, followed by the reduction of haloperidol to reduced haloperidol and by CYP-mediated oxidation. Haloperidol 62-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-171 10628896-4 1999 In studies of CYP-mediated disposition in vitro, CYP3A4 appears to be the major isoform responsible for the metabolism of haloperidol in humans. Haloperidol 122-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-17 10580224-8 1999 In this study cytokine-induced bone resorption was blocked with cimetidine and clotrimazole, which are selective inhibitors of the cytochrome P-450 IIIA family and 7-ethoxyresorufin, a nonspecific cytochrome P-450 inhibitor. Cimetidine 64-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 10580224-8 1999 In this study cytokine-induced bone resorption was blocked with cimetidine and clotrimazole, which are selective inhibitors of the cytochrome P-450 IIIA family and 7-ethoxyresorufin, a nonspecific cytochrome P-450 inhibitor. Cimetidine 64-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-213 10580224-8 1999 In this study cytokine-induced bone resorption was blocked with cimetidine and clotrimazole, which are selective inhibitors of the cytochrome P-450 IIIA family and 7-ethoxyresorufin, a nonspecific cytochrome P-450 inhibitor. Clotrimazole 79-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 10580224-8 1999 In this study cytokine-induced bone resorption was blocked with cimetidine and clotrimazole, which are selective inhibitors of the cytochrome P-450 IIIA family and 7-ethoxyresorufin, a nonspecific cytochrome P-450 inhibitor. Clotrimazole 79-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-213 10647906-13 1999 The present study suggested that the selectivity of inhibition or inactivation of human CYP isoforms by methylenedioxyphenyl compounds may vary according to the structure of the side chain. compounds 125-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 10604819-10 1999 Furthermore, knowledge of the in vivo interaction between CLB and FBM could help in identifying the CYP isoforms involved in the metabolism of both CLB and N-CLB. Clobazam 58-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 10604819-10 1999 Furthermore, knowledge of the in vivo interaction between CLB and FBM could help in identifying the CYP isoforms involved in the metabolism of both CLB and N-CLB. Felbamate 66-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 10604819-10 1999 Furthermore, knowledge of the in vivo interaction between CLB and FBM could help in identifying the CYP isoforms involved in the metabolism of both CLB and N-CLB. Clobazam 148-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 10604819-10 1999 Furthermore, knowledge of the in vivo interaction between CLB and FBM could help in identifying the CYP isoforms involved in the metabolism of both CLB and N-CLB. n-clb 156-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 10647906-2 1999 A series of methylenedioxyphenyl compounds were evaluated for their inhibitory and inactivation effects on nine human cytochrome P450 (CYP) activities using microsomes from human B-lymphoblast cells expressing specific human CYP isoforms. compounds 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-133 10630892-0 1999 Metabolism of anabolic steroids by recombinant human cytochrome P450 enzymes. Steroids 23-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 10647906-2 1999 A series of methylenedioxyphenyl compounds were evaluated for their inhibitory and inactivation effects on nine human cytochrome P450 (CYP) activities using microsomes from human B-lymphoblast cells expressing specific human CYP isoforms. compounds 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-138 10570242-0 1999 Fatty acid metabolism, conformational change, and electron transfer in cytochrome P-450(BM-3). Fatty Acids 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-92 10870095-0 1999 Inhibition of human cytochrome P450 isoforms in vitro by zafirlukast. zafirlukast 57-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 10870095-8 1999 Clinically important inhibition by zafirlukast of other CYP isoforms is not established. zafirlukast 35-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 10534319-0 1999 Cytochrome P-450-mediated metabolism of the individual enantiomers of the antidepressant agent reboxetine in human liver microsomes. Reboxetine 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 10534312-0 1999 Prediction of human liver microsomal oxidations of 7-ethoxycoumarin and chlorzoxazone with kinetic parameters of recombinant cytochrome P-450 enzymes. 7-ethoxycoumarin 51-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 10534312-0 1999 Prediction of human liver microsomal oxidations of 7-ethoxycoumarin and chlorzoxazone with kinetic parameters of recombinant cytochrome P-450 enzymes. Chlorzoxazone 72-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 10534309-0 1999 Identification of human cytochrome P-450 isoforms involved in metabolism of R(+)- and S(-)-gallopamil: utility of in vitro disappearance rate. Ranolazine 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 10534309-0 1999 Identification of human cytochrome P-450 isoforms involved in metabolism of R(+)- and S(-)-gallopamil: utility of in vitro disappearance rate. Gallopamil 86-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 10534319-1 1999 In vitro studies were conducted to identify the hepatic cytochrome P-450 (CYP) enzymes responsible for the oxidative metabolism of the individual enantiomers of reboxetine. Reboxetine 161-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 10534309-1 1999 Isoforms of cytochrome P-450 (CYP) involved in the metabolism of gallopamil enantiomers were identified by measuring the disappearance rate of parent drug from an incubation mixture with human liver microsomes and recombinant human CYPs. Gallopamil 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 10534312-1 1999 Different roles of individual forms of human cytochrome P-450 (CYP) in the oxidation of 7-ethoxycoumarin and chlorzoxazone were investigated in liver microsomes of different human samples, and the microsomal activities thus obtained were predicted with kinetic parameters obtained from cDNA-derived recombinant CYP enzymes in microsomes of Trichoplusia ni cells. 7-ethoxycoumarin 88-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 10534312-1 1999 Different roles of individual forms of human cytochrome P-450 (CYP) in the oxidation of 7-ethoxycoumarin and chlorzoxazone were investigated in liver microsomes of different human samples, and the microsomal activities thus obtained were predicted with kinetic parameters obtained from cDNA-derived recombinant CYP enzymes in microsomes of Trichoplusia ni cells. 7-ethoxycoumarin 88-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 10534309-1 1999 Isoforms of cytochrome P-450 (CYP) involved in the metabolism of gallopamil enantiomers were identified by measuring the disappearance rate of parent drug from an incubation mixture with human liver microsomes and recombinant human CYPs. Gallopamil 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 10534319-1 1999 In vitro studies were conducted to identify the hepatic cytochrome P-450 (CYP) enzymes responsible for the oxidative metabolism of the individual enantiomers of reboxetine. Reboxetine 161-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 10534309-6 1999 Among the 10 recombinant human CYP isoforms, CYP3A4 exhibited the highest CL(int) of gallopamil enantiomers, and CYP2C8 and CYP2D6 also exhibited appreciable activity. Gallopamil 85-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 10534309-10 1999 The present study suggests that CYP3A4 is a major isoform involved in the overall metabolic clearance of gallopamil enantiomers in the human liver, and that the present approach based on disappearance rate may be applicable to identify major isoforms of CYP involved in the metabolism of a drug in human liver microsomes. Gallopamil 105-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 10534312-1 1999 Different roles of individual forms of human cytochrome P-450 (CYP) in the oxidation of 7-ethoxycoumarin and chlorzoxazone were investigated in liver microsomes of different human samples, and the microsomal activities thus obtained were predicted with kinetic parameters obtained from cDNA-derived recombinant CYP enzymes in microsomes of Trichoplusia ni cells. Chlorzoxazone 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 10534312-1 1999 Different roles of individual forms of human cytochrome P-450 (CYP) in the oxidation of 7-ethoxycoumarin and chlorzoxazone were investigated in liver microsomes of different human samples, and the microsomal activities thus obtained were predicted with kinetic parameters obtained from cDNA-derived recombinant CYP enzymes in microsomes of Trichoplusia ni cells. Chlorzoxazone 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-66 10534312-2 1999 Of 14 forms of recombinant CYP examined, CYP1A1 had the highest activities (V(max)/K(m) ratio) in catalyzing 7-ethoxycoumarin O-deethylation followed by CYP1A2, 2E1, 2A6, and 2B6, although CYP1A1 has been shown to be an extrahepatic enzyme. 7-ethoxycoumarin 109-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 10534312-4 1999 Similarly, chlorzoxazone 6-hydroxylation activities of liver microsomes were predicted with kinetic parameters of recombinant human CYP enzymes and it was found that CYP3A4 as well as CYP1A2 and 2E1 were involved in chlorzoxazone hydroxylation, depending on the contents of these CYP forms in the livers. Chlorzoxazone 11-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 10534312-4 1999 Similarly, chlorzoxazone 6-hydroxylation activities of liver microsomes were predicted with kinetic parameters of recombinant human CYP enzymes and it was found that CYP3A4 as well as CYP1A2 and 2E1 were involved in chlorzoxazone hydroxylation, depending on the contents of these CYP forms in the livers. Chlorzoxazone 11-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-169 10534312-4 1999 Similarly, chlorzoxazone 6-hydroxylation activities of liver microsomes were predicted with kinetic parameters of recombinant human CYP enzymes and it was found that CYP3A4 as well as CYP1A2 and 2E1 were involved in chlorzoxazone hydroxylation, depending on the contents of these CYP forms in the livers. Chlorzoxazone 216-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 10579149-1 1999 Vesnarinone is an orally administered inotropic agent that is metabolized in vitro by the cytochrome P450 (CYP) isozymes CYP3A4 and CYP2E1. vesnarinone 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 10534312-4 1999 Similarly, chlorzoxazone 6-hydroxylation activities of liver microsomes were predicted with kinetic parameters of recombinant human CYP enzymes and it was found that CYP3A4 as well as CYP1A2 and 2E1 were involved in chlorzoxazone hydroxylation, depending on the contents of these CYP forms in the livers. Chlorzoxazone 216-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-169 10534312-5 1999 Recombinant CYP2A6 and 2B6 and CYP2D6 had considerable roles (V(max)/K(m) ratio) for 7-ethoxycoumarin O-deethylation and chlorzoxazone 6-hydroxylation, respectively; however, these CYP forms had relatively minor roles in the reactions, probably due to low expression in human livers. 7-ethoxycoumarin 85-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 10534312-5 1999 Recombinant CYP2A6 and 2B6 and CYP2D6 had considerable roles (V(max)/K(m) ratio) for 7-ethoxycoumarin O-deethylation and chlorzoxazone 6-hydroxylation, respectively; however, these CYP forms had relatively minor roles in the reactions, probably due to low expression in human livers. Chlorzoxazone 121-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 10534317-4 1999 The role of CYP enzymes in the stereoselective metabolism of R-IFA and S-IFA was investigated by monitoring the formation of both 4-hydroxy (activated) and N-dechloroethyl (DCl) (inactive, neurotoxic) metabolites. 4-hydroxyphenylacetic acid 130-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 10534317-4 1999 The role of CYP enzymes in the stereoselective metabolism of R-IFA and S-IFA was investigated by monitoring the formation of both 4-hydroxy (activated) and N-dechloroethyl (DCl) (inactive, neurotoxic) metabolites. n-dechloroethyl 156-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 10534317-4 1999 The role of CYP enzymes in the stereoselective metabolism of R-IFA and S-IFA was investigated by monitoring the formation of both 4-hydroxy (activated) and N-dechloroethyl (DCl) (inactive, neurotoxic) metabolites. dcl 173-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-15 10584978-0 1999 Effect of fusidic acid on the hepatic cytochrome P450 enzyme system. Fusidic Acid 10-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 10584978-1 1999 OBJECTIVE: To investigate the effects of fusidic acid therapy on the hepatic cytochrome P450 (CYP450) enzyme system. Fusidic Acid 41-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 77-92 10584978-1 1999 OBJECTIVE: To investigate the effects of fusidic acid therapy on the hepatic cytochrome P450 (CYP450) enzyme system. Fusidic Acid 41-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-100 10584978-9 1999 However, in contrast an activation of the CYP450 enzyme system was observed in group B after 28 days of fusidic acid therapy with an increase of total antipyrine clearance (43.0 +/- 7.62 ml/min to 51.0 +/- 9.03 ml/min) as well as clearances to all metabolites (NORA 7.11 +/- 1.75 to 8.60 +/-2.10 ml/min, OHA 11.5 +/- 2.89 to 14.0 +/- 3.97 ml/min, HMA 4.05 +/- 0.99 to 4.94 +/- 1.27 ml/min). Fusidic Acid 104-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-48 10584978-9 1999 However, in contrast an activation of the CYP450 enzyme system was observed in group B after 28 days of fusidic acid therapy with an increase of total antipyrine clearance (43.0 +/- 7.62 ml/min to 51.0 +/- 9.03 ml/min) as well as clearances to all metabolites (NORA 7.11 +/- 1.75 to 8.60 +/-2.10 ml/min, OHA 11.5 +/- 2.89 to 14.0 +/- 3.97 ml/min, HMA 4.05 +/- 0.99 to 4.94 +/- 1.27 ml/min). 10-oxohexadecanoic acid 304-307 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-48 10584978-9 1999 However, in contrast an activation of the CYP450 enzyme system was observed in group B after 28 days of fusidic acid therapy with an increase of total antipyrine clearance (43.0 +/- 7.62 ml/min to 51.0 +/- 9.03 ml/min) as well as clearances to all metabolites (NORA 7.11 +/- 1.75 to 8.60 +/-2.10 ml/min, OHA 11.5 +/- 2.89 to 14.0 +/- 3.97 ml/min, HMA 4.05 +/- 0.99 to 4.94 +/- 1.27 ml/min). 3-hydroxymethylantipyrine 347-350 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-48 10584978-11 1999 CONCLUSIONS: Our results suggest that fusidic acid has a time-dependent activating effect on the CYP450 enzyme system. Fusidic Acid 38-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-103 10575556-7 1999 Although the dogma is that the myoglobin is the source of iron, recent studies suggest that cytochrome P450 may be an important source of iron in this model. Iron 138-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 10575556-11 1999 Recent studies indicate that cytochrome P450 may also be an important source of the catalytic iron in cisplatin nephrotoxicity. Iron 94-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 10575556-11 1999 Recent studies indicate that cytochrome P450 may also be an important source of the catalytic iron in cisplatin nephrotoxicity. Cisplatin 102-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 10579149-1 1999 Vesnarinone is an orally administered inotropic agent that is metabolized in vitro by the cytochrome P450 (CYP) isozymes CYP3A4 and CYP2E1. vesnarinone 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-110 10540427-2 1999 This process is the first structural evidence for an effective method for the activation of molecular oxygen as postulated for the cytochrome P-450 system. Oxygen 102-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 10761213-0 1999 [The effect of gamma-L-glutamyl histamine on the severity of experimental anaphylactic reaction, hormonal status and liver cytochrome p-450]. gamma-l-glutamyl histamine 15-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 10574186-3 1999 Using the developed method, metabolism of coumarin in 11 samples of human liver microsomes was evaluated and 1790+/-690 pmol/min/nmol cytochrome P450 (CYP) activity was found. coumarin 42-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-149 10574186-3 1999 Using the developed method, metabolism of coumarin in 11 samples of human liver microsomes was evaluated and 1790+/-690 pmol/min/nmol cytochrome P450 (CYP) activity was found. coumarin 42-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 10655974-3 1999 Our aim was to assess the effect of dietary intake on in vivo cytochrome P450 (CYP) activities in eleven patients with abnormal liver function tests potentially due to fatty liver and associated with a high-sugar diet. Sugars 207-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 10583023-0 1999 Identification of the human cytochrome P450 enzymes involved in the in vitro metabolism of artemisinin. artemisinin 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 10583023-1 1999 AIMS: The study aimed to identify the specific human cytochrome P450 (CYP450) enzymes involved in the metabolism of artemisinin. artemisinin 116-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 10498654-0 1999 Induction of cytochrome P450 enzymes and generation of protein-aldehyde adducts are associated with sex-dependent sensitivity to alcohol-induced liver disease in micropigs. Alcohols 129-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 10583023-1 1999 AIMS: The study aimed to identify the specific human cytochrome P450 (CYP450) enzymes involved in the metabolism of artemisinin. artemisinin 116-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-76 10583023-2 1999 METHODS: Microsomes from human B-lymphoblastoid cell lines transformed with individual CYP450 cDNAs were investigated for their capacity to metabolize artemisinin. artemisinin 151-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-93 10583023-4 1999 The relative contribution of individual CYP450 isoenzymes to artemisinin metabolism in human liver microsomes was evaluated with a tree-based regression model of artemisinin disappearance rate and specific CYP450 activities. artemisinin 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-46 10498654-4 1999 Ethanol feeding increased the hepatic content of all CYP forms. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 10498654-7 1999 In the noncastrated ethanol-fed micropigs a low expression of each CYP form was associated with scant evidence of aldehyde-protein adducts. Ethanol 20-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 10498654-7 1999 In the noncastrated ethanol-fed micropigs a low expression of each CYP form was associated with scant evidence of aldehyde-protein adducts. Aldehydes 114-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 10498654-8 1999 Significant correlations emerged between the levels of different CYP forms, protein adducts, and plasma levels of sex steroids. Steroids 118-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 10511133-1 1999 Although endothelium-derived hyperpolarizing factor (EDHF) is thought to be a cytochrome P-450 product (arachidonic acid metabolite) in some tissues, in porcine coronary arteries (PCAs) its nature remains unclear. endothelium-derived hyperpolarizing factor 9-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 10511133-1 1999 Although endothelium-derived hyperpolarizing factor (EDHF) is thought to be a cytochrome P-450 product (arachidonic acid metabolite) in some tissues, in porcine coronary arteries (PCAs) its nature remains unclear. edhf 53-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 10490896-8 1999 It was concluded that lovastatin is metabolized by cytochrome P-450 3A enzymes in the small intestine. Lovastatin 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 10525052-0 1999 Renal cytochrome P450 omega-hydroxylase and epoxygenase activity are differentially modified by nitric oxide and sodium chloride. Nitric Oxide 96-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 10525052-0 1999 Renal cytochrome P450 omega-hydroxylase and epoxygenase activity are differentially modified by nitric oxide and sodium chloride. Sodium Chloride 113-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 10525052-2 1999 To probe the basis of this effect, we characterized the effects of nitric oxide (NO), a known suppressor of cytochrome P450 (CYP) enzymes, on metabolism of arachidonic acid (AA), the expression of omega-hydroxylase, and the efflux of 20-hydroxyeicosatetraenoic acid (20-HETE) from the isolated kidney. Nitric Oxide 67-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-123 10525052-2 1999 To probe the basis of this effect, we characterized the effects of nitric oxide (NO), a known suppressor of cytochrome P450 (CYP) enzymes, on metabolism of arachidonic acid (AA), the expression of omega-hydroxylase, and the efflux of 20-hydroxyeicosatetraenoic acid (20-HETE) from the isolated kidney. Nitric Oxide 67-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 10490896-9 1999 Compared with lovastatin, the cytochrome P-450-dependent intestinal intrinsic clearance of pravastatin was >5000-fold lower and cannot be expected to significantly affect its oral bioavailability or to be a significant site of drug interactions. Pravastatin 91-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. Atorvastatin 46-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 10506751-0 1999 A quantitative comparison of dibenzo[a,l]pyrene-DNA adduct formation by recombinant human cytochrome P450 microsomes. dibenzo(a,l)pyrene 29-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-105 10522495-0 1999 Effects of pretreatment with cytochrome P-450 inducers, especially phenobarbital on triphenyltin metabolism and toxicity in hamsters. triphenyltin 84-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 10522495-1 1999 The effects of cytochrome P-450 (CYP) induction by phenobarbital (PB), CYP 2B, 2C, and 3A inducer in mammalians, on triphenyltin metabolism and toxicity in hamsters were studied. Phenobarbital 51-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 10522495-1 1999 The effects of cytochrome P-450 (CYP) induction by phenobarbital (PB), CYP 2B, 2C, and 3A inducer in mammalians, on triphenyltin metabolism and toxicity in hamsters were studied. Phenobarbital 51-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 10522495-1 1999 The effects of cytochrome P-450 (CYP) induction by phenobarbital (PB), CYP 2B, 2C, and 3A inducer in mammalians, on triphenyltin metabolism and toxicity in hamsters were studied. Phenobarbital 66-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 10522495-1 1999 The effects of cytochrome P-450 (CYP) induction by phenobarbital (PB), CYP 2B, 2C, and 3A inducer in mammalians, on triphenyltin metabolism and toxicity in hamsters were studied. Phenobarbital 66-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 10522495-3 1999 Although the triphenyltin produced marked but reversible hyperglycemia and hypertriglyceridemia in PB-untreated hamsters, the pretreatment of hamsters with PB, which increased levels of CYP, suppressed the diabetogenic effects compared with PB-untreated hamsters. Phenobarbital 156-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-189 10522495-3 1999 Although the triphenyltin produced marked but reversible hyperglycemia and hypertriglyceridemia in PB-untreated hamsters, the pretreatment of hamsters with PB, which increased levels of CYP, suppressed the diabetogenic effects compared with PB-untreated hamsters. Phenobarbital 156-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-189 10522495-10 1999 These findings suggest that the induction of CYP system enzymes affects the metabolism and toxicity of triphenyltin in hamsters. triphenyltin 103-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 10522495-11 1999 Especially, based on effects of PB and other CYP inducers, PB induction has a key role in suppressing the diabetogenic action of triphenyltin, i.e. by decreasing triphenyltin accumulation in the hamsters. Phenobarbital 59-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. Lovastatin 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. Lovastatin 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-136 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. Simvastatin 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. Simvastatin 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-136 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. Atorvastatin 46-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-136 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. cerivastatin 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. cerivastatin 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-136 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. Fluvastatin 77-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 10575059-5 1999 The lipophilic drugs lovastatin, simvastatin, atorvastatin, cerivastatin and fluvastatin are metabolized via the cytochrome P450 (CYP450) system in the liver and the gut, making them subject to potential interactions with concomitantly administered drugs that are competing for metabolism via this system. Fluvastatin 77-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-136 10575059-6 1999 Clinically important interactions with simvastatin or lovastatin and drugs that inhibit the 3A4 isoenzyme (part of the CYP450 system) may result in myopathy and rhabdomyolysis, which can be fatal. Simvastatin 39-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-125 10575059-6 1999 Clinically important interactions with simvastatin or lovastatin and drugs that inhibit the 3A4 isoenzyme (part of the CYP450 system) may result in myopathy and rhabdomyolysis, which can be fatal. Lovastatin 54-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-125 10484519-1 1999 The medullary thick ascending limb (MTAL) metabolizes arachidonic acid (AA) via cytochrome P-450 (CyP450)- and cyclooxygenase (COX)-dependent pathways. Arachidonic Acid 54-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-125 10460798-1 1999 Activities of testosterone, nifedipine, and midazolam oxidation by recombinant cytochrome P-450 (P-450) 3A4 coexpressed with human NADPH-P-450 reductase (NPR) in bacterial membranes (CYP3A4/NPR membranes) were determined in comparison with those of other recombinant systems and of human liver microsomes with high contents of CYP3A4. Nifedipine 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 10583049-11 1999 These studies also indicate that as yet unknown isoforms of cytochrome P450 may be involved in RA metabolism in keratinocytes. Tretinoin 95-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 10460798-1 1999 Activities of testosterone, nifedipine, and midazolam oxidation by recombinant cytochrome P-450 (P-450) 3A4 coexpressed with human NADPH-P-450 reductase (NPR) in bacterial membranes (CYP3A4/NPR membranes) were determined in comparison with those of other recombinant systems and of human liver microsomes with high contents of CYP3A4. Testosterone 14-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 10460808-2 1999 The aim of the present study was to identify the human cytochrome P-450 (CYP) isoforms involved in zopiclone metabolism in vitro. zopiclone 99-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 10460798-1 1999 Activities of testosterone, nifedipine, and midazolam oxidation by recombinant cytochrome P-450 (P-450) 3A4 coexpressed with human NADPH-P-450 reductase (NPR) in bacterial membranes (CYP3A4/NPR membranes) were determined in comparison with those of other recombinant systems and of human liver microsomes with high contents of CYP3A4. Midazolam 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 10460808-2 1999 The aim of the present study was to identify the human cytochrome P-450 (CYP) isoforms involved in zopiclone metabolism in vitro. zopiclone 99-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 11720625-3 1999 However, clarithromycin is thought to be less frequently associated with drug induced TdP, because it inactivates hepatic cytochrome P-450 to a lesser extent than erythromycin. Clarithromycin 9-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 10548450-0 1999 Identification of human cytochrome P450 isoforms involved in the metabolism of S-2-[4-(3-methyl-2-thienyl)phenyl]propionic acid. 2-(4-(3-methyl-2-thienyl)phenyl)propionic acid 79-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 10548450-12 1999 When incubated with microsomes containing cDNA-expressed CYP isozymes, S-MTPPA was substantially oxidized to MA6 only by CYP2C9. 2-(4-(3-methyl-2-thienyl)phenyl)propionic acid 71-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-60 10917607-0 1999 Sphingomyelin hydrolysis and regulation of the expression of the gene for cytochrome P450. Sphingomyelins 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 10491726-0 1999 Review article: cytochrome P450 and the metabolism of proton pump inhibitors--emphasis on rabeprazole. Rabeprazole 90-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 10424762-7 1999 While the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P-450 (Clement et al., Biochem Pharmacol 46: 2249-2267, 1993), the resultant N-hydroxyguanidine decoupled the monooxygenase. Nitrogen 10-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 10424762-7 1999 While the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P-450 (Clement et al., Biochem Pharmacol 46: 2249-2267, 1993), the resultant N-hydroxyguanidine decoupled the monooxygenase. Guanidine 33-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 10424762-7 1999 While the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P-450 (Clement et al., Biochem Pharmacol 46: 2249-2267, 1993), the resultant N-hydroxyguanidine decoupled the monooxygenase. hydroxyguanidine 176-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 10424762-13 1999 In conclusion, similarities (formation of a urea derivative) and differences (formation of a cyanamide derivative) between the physiological oxidation of N-hydroxy-L-arginine by nitric oxide synthases and non-physiological N-hydroxyguanidines by cytochrome P-450 were observed. hydroxyguanidine 223-242 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 246-262 10421623-0 1999 In vitro identification of the human cytochrome P-450 enzymes involved in the N-demethylation of azelastine. azelastine 97-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 10421623-2 1999 The human cytochrome P-450 (CYP) isoform responsible for azelastine N-demethylation, the major metabolic pathway for azelastine, has been examined. azelastine 57-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 10421623-2 1999 The human cytochrome P-450 (CYP) isoform responsible for azelastine N-demethylation, the major metabolic pathway for azelastine, has been examined. azelastine 57-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 10421623-2 1999 The human cytochrome P-450 (CYP) isoform responsible for azelastine N-demethylation, the major metabolic pathway for azelastine, has been examined. azelastine 117-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 10421623-2 1999 The human cytochrome P-450 (CYP) isoform responsible for azelastine N-demethylation, the major metabolic pathway for azelastine, has been examined. azelastine 117-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-31 10460168-0 1999 Engineering cytochrome c peroxidase into cytochrome P450: a proximal effect on heme-thiolate ligation. heme-thiolate 79-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 10460168-1 1999 In an effort to investigate factors required to stabilize heme-thiolate ligation, key structural components necessary to convert cytochrome c peroxidase (CcP) into a thiolate-ligated cytochrome P450-like enzyme have been evaluated and the H175C/D235L CcP double mutant has been engineered. Heme 58-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-198 10460168-1 1999 In an effort to investigate factors required to stabilize heme-thiolate ligation, key structural components necessary to convert cytochrome c peroxidase (CcP) into a thiolate-ligated cytochrome P450-like enzyme have been evaluated and the H175C/D235L CcP double mutant has been engineered. thiolate 63-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-198 10460168-1 1999 In an effort to investigate factors required to stabilize heme-thiolate ligation, key structural components necessary to convert cytochrome c peroxidase (CcP) into a thiolate-ligated cytochrome P450-like enzyme have been evaluated and the H175C/D235L CcP double mutant has been engineered. thiolate 166-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-198 10411602-0 1999 Selectivity of the multidrug resistance modulator, LY335979, for P-glycoprotein and effect on cytochrome P-450 activities. zosuquidar trihydrochloride 51-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 10553719-2 1999 The cytochrome P450 (CYP)-mediated metabolism of tauromustine has been evaluated in liver and lung microsomes from various species. 1-(2-chloroethyl)-3-(2-(dimethylaminosulfonyl)ethyl)-1-nitrosourea 49-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 10419542-0 1999 The soluble guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3,-a] quinoxalin-1-one is a nonselective heme protein inhibitor of nitric oxide synthase and other cytochrome P-450 enzymes involved in nitric oxide donor bioactivation. 1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one 39-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 10419542-0 1999 The soluble guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3,-a] quinoxalin-1-one is a nonselective heme protein inhibitor of nitric oxide synthase and other cytochrome P-450 enzymes involved in nitric oxide donor bioactivation. Nitric Oxide 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 161-177 20654512-0 1999 Imipramine for Cytochrome P450 Activity Determination: a Multiple-species Metabolic Probe. Imipramine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-30 20654512-6 1999 To conclude, the metabolic profile of imipramine can be used to reveal a number of cytochrome P450 enzymes active in microsomal fractions. Imipramine 38-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-98 10553719-2 1999 The cytochrome P450 (CYP)-mediated metabolism of tauromustine has been evaluated in liver and lung microsomes from various species. 1-(2-chloroethyl)-3-(2-(dimethylaminosulfonyl)ethyl)-1-nitrosourea 49-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 10455289-8 1999 In contrast, the selective CYT-P450 hydroxylase inhibitor, N-methylsulphonyl-12,12-dibromododec-11-enamide (DDMS) attenuated the vascular response to increasing renal perfusion pressure. n-methylsulphonyl-12,12-dibromododec-11-enamide 59-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-35 10553719-15 1999 Among the human cDNA expressed CYP enzymes, not only was tauromustine determined to be catalysed predominantly by CYP3A4, but also to some extent by CYP2C19 and CYP2D6. 1-(2-chloroethyl)-3-(2-(dimethylaminosulfonyl)ethyl)-1-nitrosourea 57-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 10409674-2 1999 Cytochrome P450 4F3 (CYP4F3) catalyzes the inactivation of leukotriene B(4) by omega-oxidation in human neutrophils. Leukotriene B4 59-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-19 10409674-2 1999 Cytochrome P450 4F3 (CYP4F3) catalyzes the inactivation of leukotriene B(4) by omega-oxidation in human neutrophils. Leukotriene B4 59-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-27 10424895-0 1999 Cytochrome P450-dependent metabolism of arachidonic acid. Arachidonic Acid 40-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10424895-1 1999 The cytochrome P450-dependent metabolism of arachidonic acid, the mechanisms of regulation of stereo- and regiospecificity by cytochrome P450 isoenzymes, and the biological relevance of metabolites of the arachidonic acid cascade is discussed in this review. Arachidonic Acid 44-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 10424895-1 1999 The cytochrome P450-dependent metabolism of arachidonic acid, the mechanisms of regulation of stereo- and regiospecificity by cytochrome P450 isoenzymes, and the biological relevance of metabolites of the arachidonic acid cascade is discussed in this review. Arachidonic Acid 205-221 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 10424895-1 1999 The cytochrome P450-dependent metabolism of arachidonic acid, the mechanisms of regulation of stereo- and regiospecificity by cytochrome P450 isoenzymes, and the biological relevance of metabolites of the arachidonic acid cascade is discussed in this review. Arachidonic Acid 205-221 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-141 10455289-8 1999 In contrast, the selective CYT-P450 hydroxylase inhibitor, N-methylsulphonyl-12,12-dibromododec-11-enamide (DDMS) attenuated the vascular response to increasing renal perfusion pressure. DDMS 108-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-35 10383930-6 1999 Oxidative metabolism of molinate in both rats and humans appears to be mediated by cytochrome P-450 and not flavin monooxygenases as indicated by the use of metabolic inhibitors. molinate 24-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 10383917-0 1999 Sertraline N-demethylation is catalyzed by multiple isoforms of human cytochrome P-450 in vitro. Sertraline 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 10383917-2 1999 We identified the cytochrome P-450 (CYP) isoforms involved in sertraline N-demethylation using pooled human liver microsomes and cDNA-expressed CYP isoforms. Sertraline 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 10383922-0 1999 Inhibitory effects of azelastine and its metabolites on drug oxidation catalyzed by human cytochrome P-450 enzymes. azelastine 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 10383922-2 1999 In the present study, the inhibitory effects of azelastine and its two metabolites on human cytochrome P-450 (CYP) isoform-dependent reactions were investigated to predict the drug interactions of azelastine using microsomes from human B-lymphoblast cells expressing CYP. azelastine 48-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 10383922-2 1999 In the present study, the inhibitory effects of azelastine and its two metabolites on human cytochrome P-450 (CYP) isoform-dependent reactions were investigated to predict the drug interactions of azelastine using microsomes from human B-lymphoblast cells expressing CYP. azelastine 48-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. Clozapine 48-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-273 10383922-2 1999 In the present study, the inhibitory effects of azelastine and its two metabolites on human cytochrome P-450 (CYP) isoform-dependent reactions were investigated to predict the drug interactions of azelastine using microsomes from human B-lymphoblast cells expressing CYP. azelastine 197-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 10383922-2 1999 In the present study, the inhibitory effects of azelastine and its two metabolites on human cytochrome P-450 (CYP) isoform-dependent reactions were investigated to predict the drug interactions of azelastine using microsomes from human B-lymphoblast cells expressing CYP. azelastine 197-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 10381752-1 1999 In humans, meloxicam is metabolized mainly by cytochrome P-450 (CYP)-dependent hydroxylation of the 5"-methyl group. Meloxicam 11-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 10381752-1 1999 In humans, meloxicam is metabolized mainly by cytochrome P-450 (CYP)-dependent hydroxylation of the 5"-methyl group. Meloxicam 11-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 10381752-6 1999 Using expressed cytochrome P-450 enzymes and selective chemical inhibitors of CYP 2C9 and CYP 3A4, it was found that quinidine markedly increased the rate of CYP 3A4-mediated meloxicam hydroxylation but was virtually without effect on CYP 2C9. Quinidine 117-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. Clozapine 48-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 275-278 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. Risperidone 59-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-273 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. Risperidone 59-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 275-278 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. Olanzapine 72-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-273 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. Olanzapine 72-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 275-278 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. Quetiapine Fumarate 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-273 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. Quetiapine Fumarate 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 275-278 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. sertindole 99-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 258-273 10422890-1 1999 The introduction of the atypical antipsychotics clozapine, risperidone, olanzapine, quetiapine and sertindole for the treatment of schizophrenia has coincided with an increased awareness of the potential of drug-drug interactions, particularly involving the cytochrome P450 (CYP) enzymes. sertindole 99-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 275-278 10330055-3 1999 The inhibition by ANG II was abolished by pretreatment with eicosatetraynoic acid (10 microM), a general inhibitor of arachidonic acid metabolism, or 17-octadecynoic acid (10 microM), a highly selective inhibitor of cytochrome P-450 pathways. 17-octadecynoic acid 150-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-232 10348794-1 1999 The contributions of specific human liver cytochrome P-450 (CYP) enzymes to the activation, via 4-hydroxylation, of the oxazaphosphorine anticancer prodrugs cyclophosphamide (CPA) and ifosfamide (IFA) were investigated. oxazaphosphorine 120-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 10348794-1 1999 The contributions of specific human liver cytochrome P-450 (CYP) enzymes to the activation, via 4-hydroxylation, of the oxazaphosphorine anticancer prodrugs cyclophosphamide (CPA) and ifosfamide (IFA) were investigated. oxazaphosphorine 120-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 10348794-1 1999 The contributions of specific human liver cytochrome P-450 (CYP) enzymes to the activation, via 4-hydroxylation, of the oxazaphosphorine anticancer prodrugs cyclophosphamide (CPA) and ifosfamide (IFA) were investigated. Cyclophosphamide 157-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 10348794-1 1999 The contributions of specific human liver cytochrome P-450 (CYP) enzymes to the activation, via 4-hydroxylation, of the oxazaphosphorine anticancer prodrugs cyclophosphamide (CPA) and ifosfamide (IFA) were investigated. Cyclophosphamide 157-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 10348794-1 1999 The contributions of specific human liver cytochrome P-450 (CYP) enzymes to the activation, via 4-hydroxylation, of the oxazaphosphorine anticancer prodrugs cyclophosphamide (CPA) and ifosfamide (IFA) were investigated. Cyclophosphamide 175-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 10348794-1 1999 The contributions of specific human liver cytochrome P-450 (CYP) enzymes to the activation, via 4-hydroxylation, of the oxazaphosphorine anticancer prodrugs cyclophosphamide (CPA) and ifosfamide (IFA) were investigated. Cyclophosphamide 175-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 10348794-1 1999 The contributions of specific human liver cytochrome P-450 (CYP) enzymes to the activation, via 4-hydroxylation, of the oxazaphosphorine anticancer prodrugs cyclophosphamide (CPA) and ifosfamide (IFA) were investigated. Ifosfamide 184-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 10348794-1 1999 The contributions of specific human liver cytochrome P-450 (CYP) enzymes to the activation, via 4-hydroxylation, of the oxazaphosphorine anticancer prodrugs cyclophosphamide (CPA) and ifosfamide (IFA) were investigated. Ifosfamide 184-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 10348794-1 1999 The contributions of specific human liver cytochrome P-450 (CYP) enzymes to the activation, via 4-hydroxylation, of the oxazaphosphorine anticancer prodrugs cyclophosphamide (CPA) and ifosfamide (IFA) were investigated. Ifosfamide 196-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-63 10220485-2 1999 Pulegone is metabolized by human liver cytochrome P-450s to menthofuran, a proximate hepatotoxic metabolite of pulegone. pulegone 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 10220485-2 1999 Pulegone is metabolized by human liver cytochrome P-450s to menthofuran, a proximate hepatotoxic metabolite of pulegone. menthofuran 60-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 10220485-2 1999 Pulegone is metabolized by human liver cytochrome P-450s to menthofuran, a proximate hepatotoxic metabolite of pulegone. pulegone 111-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 10215634-0 1999 Cytochrome P-450-dependent bioactivation of 1,1-dichloroethylene to a reactive epoxide in human lung and liver microsomes. vinylidene chloride 44-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 10215634-0 1999 Cytochrome P-450-dependent bioactivation of 1,1-dichloroethylene to a reactive epoxide in human lung and liver microsomes. Epoxy Compounds 79-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 10215634-1 1999 We investigated the cytochrome P-450-dependent metabolism of 1, 1-dichloroethylene (DCE) by human lung and liver microsomes and compared the results from analogous experiments in mice. vinylidene chloride 61-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 10215634-1 1999 We investigated the cytochrome P-450-dependent metabolism of 1, 1-dichloroethylene (DCE) by human lung and liver microsomes and compared the results from analogous experiments in mice. ethylene dichloride 84-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 10499085-1 1999 Photoexcited iron porphyrins can be used to mimic the catalytic activity of cytochrome P-450 oxygenases both in the reduction of halogenated alkanes and in the oxidation of hydrocarbons by O2 itself at room temperature and atmospheric pressure. Oxygen 189-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 10216279-2 1999 Fourteen recombinant expressed cytochrome P450 (CYP) enzymes, human liver microsomes from different donors, and selective cytochrome P450 inhibitors were used to study the hydroxylation of taurochenodeoxycholic acid and lithocholic acid. Taurochenodeoxycholic Acid 189-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 10215728-1 1999 BACKGROUND: The S-mephenytoin hydroxylase is a polymorphic cytochrome P450 (CYP) enzyme, identified as CYP2C19, which catalyses the metabolism of omeprazole and some other drugs. Omeprazole 146-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 10215728-1 1999 BACKGROUND: The S-mephenytoin hydroxylase is a polymorphic cytochrome P450 (CYP) enzyme, identified as CYP2C19, which catalyses the metabolism of omeprazole and some other drugs. Omeprazole 146-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-79 10197604-0 1999 Comparison of cytochrome P450- and peroxidase-dependent metabolic activation of the potent carcinogen dibenzo[a,l]pyrene in human cell lines: formation of stable DNA adducts and absence of a detectable increase in apurinic sites. dibenzo(a,l)pyrene 102-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-45 10380060-5 1999 Phenytoin, phenobarbiral, primidone and carbamazepine induce CYP and UDPGT enzymes while valproic acid inhibits them. Phenytoin 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 10385214-5 1999 Formation of N-debutylhalofantrine was cytochrome P450 (CYP)-mediated. 1,3-dichloro-6-trifluoromethyl-9-phenanthryl-3-(n-butyl)aminopropanol 13-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 10380060-5 1999 Phenytoin, phenobarbiral, primidone and carbamazepine induce CYP and UDPGT enzymes while valproic acid inhibits them. phenobarbiral 11-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 10380060-5 1999 Phenytoin, phenobarbiral, primidone and carbamazepine induce CYP and UDPGT enzymes while valproic acid inhibits them. Primidone 26-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 10380060-5 1999 Phenytoin, phenobarbiral, primidone and carbamazepine induce CYP and UDPGT enzymes while valproic acid inhibits them. Carbamazepine 40-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 10499085-1 1999 Photoexcited iron porphyrins can be used to mimic the catalytic activity of cytochrome P-450 oxygenases both in the reduction of halogenated alkanes and in the oxidation of hydrocarbons by O2 itself at room temperature and atmospheric pressure. iron porphyrins 13-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 10499085-1 1999 Photoexcited iron porphyrins can be used to mimic the catalytic activity of cytochrome P-450 oxygenases both in the reduction of halogenated alkanes and in the oxidation of hydrocarbons by O2 itself at room temperature and atmospheric pressure. Alkanes 141-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 10499085-1 1999 Photoexcited iron porphyrins can be used to mimic the catalytic activity of cytochrome P-450 oxygenases both in the reduction of halogenated alkanes and in the oxidation of hydrocarbons by O2 itself at room temperature and atmospheric pressure. Hydrocarbons 173-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 10385214-5 1999 Formation of N-debutylhalofantrine was cytochrome P450 (CYP)-mediated. 1,3-dichloro-6-trifluoromethyl-9-phenanthryl-3-(n-butyl)aminopropanol 13-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-59 10385214-6 1999 Studies using selective inhibitors of individual CYPs revealed the role of CYP 3As in the formation of N-debutylhalofantrine. 1,3-dichloro-6-trifluoromethyl-9-phenanthryl-3-(n-butyl)aminopropanol 103-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 10231734-0 1999 Involvement of cytochrome P-450 isozymes in melatonin metabolism and clinical implications. Melatonin 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 10385214-8 1999 In microsomes from 12 livers from man the rate of N-debutylation of halofantrine correlated strongly with CYP 3A4 relative levels (P = 0.002) and less strongly, but significantly, with CYP 2C8 levels (P = 0.025). Nitrogen 50-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 10385214-8 1999 In microsomes from 12 livers from man the rate of N-debutylation of halofantrine correlated strongly with CYP 3A4 relative levels (P = 0.002) and less strongly, but significantly, with CYP 2C8 levels (P = 0.025). halofantrine 68-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 10385214-9 1999 To characterize CYP-mediated metabolism of halofantrine further, incubations were performed with yeast microsomes expressing specific CYP 3A4, CYP 3A5, CYP 2D6, CYP 2C8 and CYP 2C19 from man. halofantrine 43-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 10385214-9 1999 To characterize CYP-mediated metabolism of halofantrine further, incubations were performed with yeast microsomes expressing specific CYP 3A4, CYP 3A5, CYP 2D6, CYP 2C8 and CYP 2C19 from man. halofantrine 43-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-137 10385214-9 1999 To characterize CYP-mediated metabolism of halofantrine further, incubations were performed with yeast microsomes expressing specific CYP 3A4, CYP 3A5, CYP 2D6, CYP 2C8 and CYP 2C19 from man. halofantrine 43-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-137 10385214-9 1999 To characterize CYP-mediated metabolism of halofantrine further, incubations were performed with yeast microsomes expressing specific CYP 3A4, CYP 3A5, CYP 2D6, CYP 2C8 and CYP 2C19 from man. halofantrine 43-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-137 10385214-9 1999 To characterize CYP-mediated metabolism of halofantrine further, incubations were performed with yeast microsomes expressing specific CYP 3A4, CYP 3A5, CYP 2D6, CYP 2C8 and CYP 2C19 from man. halofantrine 43-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-137 10209701-6 1999 Concomitant administration with drugs that inhibit the hepatic cytochrome P-450 (imidazole antifungals, macrolide antibiotics) or those that prolong the QT interval by the same or other mechanism (e.g. antiarrhythmics, antipsychotics, tricyclic antidepressants) increases their effect on the cardiac repolarization. imidazole 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 11670903-0 1999 Role of the Invariant Peptide Fragment Forming NH.S Hydrogen Bonds in the Active Site of Cytochrome P-450 and Chloroperoxidase: Synthesis and Properties of Cys-Containing Peptide Fe(III) and Ga(III) (Octaethylporphinato) Complexes as Models. Hydrogen 52-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-126 11670903-0 1999 Role of the Invariant Peptide Fragment Forming NH.S Hydrogen Bonds in the Active Site of Cytochrome P-450 and Chloroperoxidase: Synthesis and Properties of Cys-Containing Peptide Fe(III) and Ga(III) (Octaethylporphinato) Complexes as Models. Cysteine 156-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-126 10209701-6 1999 Concomitant administration with drugs that inhibit the hepatic cytochrome P-450 (imidazole antifungals, macrolide antibiotics) or those that prolong the QT interval by the same or other mechanism (e.g. antiarrhythmics, antipsychotics, tricyclic antidepressants) increases their effect on the cardiac repolarization. macrolide antibiotics 104-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 10084413-7 1999 Miconazole exerts its fungistatic action by inhibiting some isoenzymes of the fungal cytochrome P450 system. Miconazole 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 10064574-0 1999 The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor fluvastatin: effect on human cytochrome P-450 and implications for metabolic drug interactions. Fluvastatin 62-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 10064574-12 1999 This data indicates that several human cytochrome P-450 enzymes metabolize fluvastatin with CYP2C9 contributing 50-80%. Fluvastatin 75-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 10980926-4 1999 This liver injury is due not to the drug itself but to the formation of the toxic metabolite N-acetyl-p-benzoquinine imine generated through the cytochrome P-450 drug-metabolizing system. n-acetyl-p-benzoquinine imine 93-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-161 9929520-0 1999 Cytochrome P-450 isoforms involved in carboxylic acid ester cleavage of Hantzsch pyridine ester of pranidipine. carboxylic acid ester 38-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 9929518-3 1999 Irbesartan oxidation follows Michaelis-Menten kinetics, consistent with the involvement of a single CYP isoform in these hydroxylation processes. Irbesartan 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-103 9929499-0 1999 Comparison of cytochrome P-450-dependent metabolism and drug interactions of the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors lovastatin and pravastatin in the liver. Lovastatin 133-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 9929499-0 1999 Comparison of cytochrome P-450-dependent metabolism and drug interactions of the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors lovastatin and pravastatin in the liver. Pravastatin 148-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 9929518-8 1999 Oxidation of irbesartan was markedly inhibited by sulfaphenazole (CYP2C9 inhibitor), but not by any of several other CYP inhibitors. Irbesartan 13-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 9929520-0 1999 Cytochrome P-450 isoforms involved in carboxylic acid ester cleavage of Hantzsch pyridine ester of pranidipine. hantzsch pyridine ester 72-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 9929518-8 1999 Oxidation of irbesartan was markedly inhibited by sulfaphenazole (CYP2C9 inhibitor), but not by any of several other CYP inhibitors. Sulfaphenazole 50-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 9929520-0 1999 Cytochrome P-450 isoforms involved in carboxylic acid ester cleavage of Hantzsch pyridine ester of pranidipine. pranidipine 99-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 9929520-1 1999 Cytochrome P-450 (CYP) isoforms responsible for the cleavage of Hantzsch pyridine ester at the 3-position of pranidipine were studied in vitro using cDNA-expressed human CYP enzymes. hantzsch pyridine ester 64-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 9929520-1 1999 Cytochrome P-450 (CYP) isoforms responsible for the cleavage of Hantzsch pyridine ester at the 3-position of pranidipine were studied in vitro using cDNA-expressed human CYP enzymes. hantzsch pyridine ester 64-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 9929520-1 1999 Cytochrome P-450 (CYP) isoforms responsible for the cleavage of Hantzsch pyridine ester at the 3-position of pranidipine were studied in vitro using cDNA-expressed human CYP enzymes. pranidipine 109-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 9929520-1 1999 Cytochrome P-450 (CYP) isoforms responsible for the cleavage of Hantzsch pyridine ester at the 3-position of pranidipine were studied in vitro using cDNA-expressed human CYP enzymes. pranidipine 109-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 10048600-3 1999 In the present study, in vitro metabolism of risperidone (100 microM) was investigated using the recombinant human cytochrome P450 (CYP) enzymes CYP1A1, CYP1A2, CYP2C8, CYP2C9-arg144, CYP2C9-cys144, CYP2C19, CYP2D6, CYP3A4 and CYP3A5 supplemented with an NADPH-generating system. Risperidone 45-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-130 10563966-2 1999 Cytochrome P450 (CP450) catalyzed oxidative metabolism of carbofuran (1), carbaryl (2), and pirimicarb (3) has been modeled using biomimetic oxidations catalyzed by iron(III) tetraarylporphyrins. Carbofuran 58-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10563966-2 1999 Cytochrome P450 (CP450) catalyzed oxidative metabolism of carbofuran (1), carbaryl (2), and pirimicarb (3) has been modeled using biomimetic oxidations catalyzed by iron(III) tetraarylporphyrins. Carbofuran 58-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-22 10563966-2 1999 Cytochrome P450 (CP450) catalyzed oxidative metabolism of carbofuran (1), carbaryl (2), and pirimicarb (3) has been modeled using biomimetic oxidations catalyzed by iron(III) tetraarylporphyrins. Carbaryl 74-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10563966-2 1999 Cytochrome P450 (CP450) catalyzed oxidative metabolism of carbofuran (1), carbaryl (2), and pirimicarb (3) has been modeled using biomimetic oxidations catalyzed by iron(III) tetraarylporphyrins. Carbaryl 74-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-22 10563966-2 1999 Cytochrome P450 (CP450) catalyzed oxidative metabolism of carbofuran (1), carbaryl (2), and pirimicarb (3) has been modeled using biomimetic oxidations catalyzed by iron(III) tetraarylporphyrins. pirimicarb 92-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10563966-2 1999 Cytochrome P450 (CP450) catalyzed oxidative metabolism of carbofuran (1), carbaryl (2), and pirimicarb (3) has been modeled using biomimetic oxidations catalyzed by iron(III) tetraarylporphyrins. pirimicarb 92-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-22 10563966-2 1999 Cytochrome P450 (CP450) catalyzed oxidative metabolism of carbofuran (1), carbaryl (2), and pirimicarb (3) has been modeled using biomimetic oxidations catalyzed by iron(III) tetraarylporphyrins. iron(iii) tetraarylporphyrins 165-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 10563966-2 1999 Cytochrome P450 (CP450) catalyzed oxidative metabolism of carbofuran (1), carbaryl (2), and pirimicarb (3) has been modeled using biomimetic oxidations catalyzed by iron(III) tetraarylporphyrins. iron(iii) tetraarylporphyrins 165-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-22 10048600-3 1999 In the present study, in vitro metabolism of risperidone (100 microM) was investigated using the recombinant human cytochrome P450 (CYP) enzymes CYP1A1, CYP1A2, CYP2C8, CYP2C9-arg144, CYP2C9-cys144, CYP2C19, CYP2D6, CYP3A4 and CYP3A5 supplemented with an NADPH-generating system. Risperidone 45-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 10048600-3 1999 In the present study, in vitro metabolism of risperidone (100 microM) was investigated using the recombinant human cytochrome P450 (CYP) enzymes CYP1A1, CYP1A2, CYP2C8, CYP2C9-arg144, CYP2C9-cys144, CYP2C19, CYP2D6, CYP3A4 and CYP3A5 supplemented with an NADPH-generating system. NADP 255-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 10649851-3 1999 MAIN PURPOSE: In this study possible participation of eicosanoids, cytochrome P-450 and reactive oxygen species in the changes of airways reactivity evoked by toluene exposure as the source of free radicals was followed up by an indirect method. Toluene 159-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 9935275-9 1999 Increased macrolide use in children in recent years has resulted in a growing potential for drug interactions between them and other pharmacologically active agents via the inhibition of cytochrome P450 (CYP) microsomal enzymes. Macrolides 10-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 187-202 10702887-1 1999 OBJECTIVE: In vitro results suggest that cilostazol is metabolised by cytochrome P450 (CYP) isoforms 1A2, 2D6, 3A4 and 2C19. Cilostazol 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 10702887-1 1999 OBJECTIVE: In vitro results suggest that cilostazol is metabolised by cytochrome P450 (CYP) isoforms 1A2, 2D6, 3A4 and 2C19. Cilostazol 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 9884321-1 1999 Biotransformation of the M1-muscarinic agonist Lu 25-109 (5-(2-ethyl-2H-tetrazol-5-yl)-1-methyl-1,2,3,6-tetrahydropyridine) , in development for the treatment of Alzheimer"s disease, was investigated to obtain information on the identity of human hepatic cytochrome P-450 enzymes involved in its metabolism. LU 25-109-T 47-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 255-271 9935275-9 1999 Increased macrolide use in children in recent years has resulted in a growing potential for drug interactions between them and other pharmacologically active agents via the inhibition of cytochrome P450 (CYP) microsomal enzymes. Macrolides 10-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-207 9935275-11 1999 Clarithromycin is a weak inducer of CYP and exhibits fewer drug-drug interactions than erythromycin. Clarithromycin 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 9872399-1 1998 Light absorption photometry of HepG2 cells treated with phenobarbital for enhancing the content of cytochrome P-450 and the synthesis of erythropoietin revealed an influence on all cytochromes detectable in the wavelength range between 400 and 620 nm. Phenobarbital 56-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 9987703-1 1999 The single-dose effects of the cytochrome P-450 inhibitors erythromycin and ketoconazole on the steady-state pharmacokinetics and electrocardiographic repolarization pharmacodynamics of intranasal levocabastine, a potent and selective H1-receptor antagonist, were evaluated in healthy young male subjects. Erythromycin 59-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 9987703-1 1999 The single-dose effects of the cytochrome P-450 inhibitors erythromycin and ketoconazole on the steady-state pharmacokinetics and electrocardiographic repolarization pharmacodynamics of intranasal levocabastine, a potent and selective H1-receptor antagonist, were evaluated in healthy young male subjects. levocabastine 197-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 9987703-10 1999 Since levocabastine undergoes only minimal hepatic metabolism and is not a substrate for or an inhibitor of cytochrome P-450, the likelihood of systemic drug interactions with drugs affecting the cytochrome P-450 system is minimal. levocabastine 6-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 196-212 10343314-9 1999 On the other hand, it must be noted that with the exception of the cytochrome P-450 metabolites of arachidonic acid, most of the major eicosanoids that are biologically active and are known to play regulatory roles in physiology and/or pathology, have commercially available antibodies and enzyme-tracers. Arachidonic Acid 99-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 10221637-6 1999 Methylphenidate appeared to be involved primarily in pharmacokinetic interactions suggestive of cytochrome P450 inhibition while dextroamphetamine and pemoline were more often involved in apparent pharmacodynamic interactions. Methylphenidate 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-111 9836023-3 1998 The present experiments were designed to identify the CYP isoenzymes involved in the metabolism of mCPP to its main metabolite p-hydroxy-mCPP (OH-mCPP). 1-(3-chlorophenyl)piperazine 99-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 9866708-0 1998 Analysis of cytochrome P450 metabolites of arachidonic and linoleic acids by liquid chromatography-mass spectrometry with ion trap MS. We have used reversed phase-high performance liquid chromatography-mass spectrometry (RP-HPLC-MS) with an ion trap mass spectrometer to study the metabolism of arachidonic and linoleic acids by human recombinant cytochrome P450 (CYP) enzymes. arachidonic 43-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 9866708-0 1998 Analysis of cytochrome P450 metabolites of arachidonic and linoleic acids by liquid chromatography-mass spectrometry with ion trap MS. We have used reversed phase-high performance liquid chromatography-mass spectrometry (RP-HPLC-MS) with an ion trap mass spectrometer to study the metabolism of arachidonic and linoleic acids by human recombinant cytochrome P450 (CYP) enzymes. Linoleic Acids 59-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 9866708-0 1998 Analysis of cytochrome P450 metabolites of arachidonic and linoleic acids by liquid chromatography-mass spectrometry with ion trap MS. We have used reversed phase-high performance liquid chromatography-mass spectrometry (RP-HPLC-MS) with an ion trap mass spectrometer to study the metabolism of arachidonic and linoleic acids by human recombinant cytochrome P450 (CYP) enzymes. arachidonic 295-306 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 9866708-0 1998 Analysis of cytochrome P450 metabolites of arachidonic and linoleic acids by liquid chromatography-mass spectrometry with ion trap MS. We have used reversed phase-high performance liquid chromatography-mass spectrometry (RP-HPLC-MS) with an ion trap mass spectrometer to study the metabolism of arachidonic and linoleic acids by human recombinant cytochrome P450 (CYP) enzymes. Linoleic Acids 311-325 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-27 9866708-9 1998 RP-HPLC-MS with MS2 seems to be useful for rapid identification of fatty acid metabolites in complex mixtures formed by cytochrome P450. Fatty Acids 67-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-135 9836023-3 1998 The present experiments were designed to identify the CYP isoenzymes involved in the metabolism of mCPP to its main metabolite p-hydroxy-mCPP (OH-mCPP). p-hydroxy-mcpp 127-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 9836023-3 1998 The present experiments were designed to identify the CYP isoenzymes involved in the metabolism of mCPP to its main metabolite p-hydroxy-mCPP (OH-mCPP). oh-mcpp 143-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-57 9836023-4 1998 METHODS: The rate of production of OH-mCPP from mCPP was correlated with isoform activities in a panel of human liver microsomes, was assessed using a panel of individual complementary DNA-expressed human CYP isoenzymes, and was investigated in the presence of a specific inhibitor of CYP2D6. oh-mcpp 35-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-208 9836023-4 1998 METHODS: The rate of production of OH-mCPP from mCPP was correlated with isoform activities in a panel of human liver microsomes, was assessed using a panel of individual complementary DNA-expressed human CYP isoenzymes, and was investigated in the presence of a specific inhibitor of CYP2D6. 1-(3-chlorophenyl)piperazine 38-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-208 9820175-5 1998 Specific substrates of CYP included testosterone (catalysed by CYP3A4), paclitaxel (CYP2C8), 7-ethoxyresorufin (CYP1A1, CYP1A2), coumarin (CYP2A6), aniline (CYP2E1) and (+/-)-bufuralol (CYP2D6). bufuralol 169-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. Rifampin 12-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. Rifampin 12-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. inn 22-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. inn 22-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. Rifampin 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 9871429-1 1998 BACKGROUND: Rifampin (INN, rifampicin) is a potent inducer of cytochrome P450 (CYP) enzymes involved in drug metabolism and therefore causes many drug interactions. Rifampin 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-82 9918136-5 1998 Adduct formation in stimulated neutrophils was inhibited 80% by the myeloperoxidase inhibitor sodium azide (1 mM) but was not affected by proadifen (100 microM), indomethacin (100 microM), or eicosatetraynoic acid (100 microM), inhibitors of cytochrome P450, prostaglandin synthetase, and lipoxygenase, respectively. Sodium Azide 94-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 242-301 9815106-2 1998 Liver cytochrome P-450 enzymes also catalyze the oxidative cleavage of C==N bonds of compounds containing a -C(NH2)==NOH function, producing NO in vitro. 2-amino-5-chlorobenzophenone N-hydroxyamidinohydrazone 117-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-22 9815106-6 1998 Ethoxyresorufin (10(-7) to 10(-4) M), an alternate cytochrome P-450 substrate, inhibited formamidoxime-induced cGMP accumulation as well as tracheal ring relaxation in cultured tracheal smooth muscle cells. ethoxyresorufin 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 9815106-6 1998 Ethoxyresorufin (10(-7) to 10(-4) M), an alternate cytochrome P-450 substrate, inhibited formamidoxime-induced cGMP accumulation as well as tracheal ring relaxation in cultured tracheal smooth muscle cells. formamidoxime 89-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 9815106-8 1998 These results suggest that NO can be synthesized from formamidoxime in tracheal smooth muscle cells, presumably by a reaction catalyzed by cytochrome P-450. formamidoxime 54-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 9820175-0 1998 Potent and non-specific inhibition of cytochrome P450 by JM216, a new oral platinum agent. satraplatin 57-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 9820175-0 1998 Potent and non-specific inhibition of cytochrome P450 by JM216, a new oral platinum agent. Platinum 75-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-53 9820175-4 1998 In this study, the capacity of JM216 to inhibit cytochrome P450 (CYP) in human liver microsomes was investigated by measuring the inhibition potential (IC50 and Ki) on prototype reactions. satraplatin 31-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 9820175-4 1998 In this study, the capacity of JM216 to inhibit cytochrome P450 (CYP) in human liver microsomes was investigated by measuring the inhibition potential (IC50 and Ki) on prototype reactions. satraplatin 31-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-68 9820175-5 1998 Specific substrates of CYP included testosterone (catalysed by CYP3A4), paclitaxel (CYP2C8), 7-ethoxyresorufin (CYP1A1, CYP1A2), coumarin (CYP2A6), aniline (CYP2E1) and (+/-)-bufuralol (CYP2D6). Testosterone 36-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 9820175-5 1998 Specific substrates of CYP included testosterone (catalysed by CYP3A4), paclitaxel (CYP2C8), 7-ethoxyresorufin (CYP1A1, CYP1A2), coumarin (CYP2A6), aniline (CYP2E1) and (+/-)-bufuralol (CYP2D6). Paclitaxel 72-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 9820175-5 1998 Specific substrates of CYP included testosterone (catalysed by CYP3A4), paclitaxel (CYP2C8), 7-ethoxyresorufin (CYP1A1, CYP1A2), coumarin (CYP2A6), aniline (CYP2E1) and (+/-)-bufuralol (CYP2D6). ethoxyresorufin 93-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 9820175-5 1998 Specific substrates of CYP included testosterone (catalysed by CYP3A4), paclitaxel (CYP2C8), 7-ethoxyresorufin (CYP1A1, CYP1A2), coumarin (CYP2A6), aniline (CYP2E1) and (+/-)-bufuralol (CYP2D6). coumarin 129-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 9820175-5 1998 Specific substrates of CYP included testosterone (catalysed by CYP3A4), paclitaxel (CYP2C8), 7-ethoxyresorufin (CYP1A1, CYP1A2), coumarin (CYP2A6), aniline (CYP2E1) and (+/-)-bufuralol (CYP2D6). aniline 148-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-26 9799565-7 1998 Upon reconstitution with P450 reductase and phospholipid, CYP4F2 converted LTB4 to 20-OH LTB4 at a turnover rate of 392 pmol/min/nmol P450, whereas the other human liver P450s tested, including CYP4A11, exhibited neglible LTB4 omega-hydroxylase activity. 20-oh ltb4 83-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-244 9820175-6 1998 JM216 inhibited the catalytic activities of CYP isozymes. satraplatin 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 9825831-2 1998 To determine which cytochrome P450 is involved in taxotere biotransformation, 11 cDNA-expressed human cytochrome P450s were examined for their activity in the metabolism of taxotere and its derivatives. Docetaxel 50-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 9790080-6 1998 The MFO activities associated with different cytochrome P-450 isoenzymes decreased to different extents in the presence of bile with the O-dealkylation of pentoxyresorufin being more labile than that of ethoxyresorufin. pentoxyresorufin 155-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 9790080-6 1998 The MFO activities associated with different cytochrome P-450 isoenzymes decreased to different extents in the presence of bile with the O-dealkylation of pentoxyresorufin being more labile than that of ethoxyresorufin. ethoxyresorufin 203-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 9763401-1 1998 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]ABT-761 and its N-dehydroxylated metabolite, [14C]ABT-438, by human liver microsomes. Carbon-14 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 9763401-1 1998 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]ABT-761 and its N-dehydroxylated metabolite, [14C]ABT-438, by human liver microsomes. Carbon-14 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 9763401-1 1998 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]ABT-761 and its N-dehydroxylated metabolite, [14C]ABT-438, by human liver microsomes. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 129-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 9763401-1 1998 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]ABT-761 and its N-dehydroxylated metabolite, [14C]ABT-438, by human liver microsomes. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 129-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 9763401-1 1998 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]ABT-761 and its N-dehydroxylated metabolite, [14C]ABT-438, by human liver microsomes. Carbon-14 175-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 9763401-1 1998 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]ABT-761 and its N-dehydroxylated metabolite, [14C]ABT-438, by human liver microsomes. Carbon-14 175-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 9763401-1 1998 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]ABT-761 and its N-dehydroxylated metabolite, [14C]ABT-438, by human liver microsomes. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 179-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 9763401-1 1998 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]ABT-761 and its N-dehydroxylated metabolite, [14C]ABT-438, by human liver microsomes. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 179-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 9844795-0 1998 Alcohol-associated rhabdomyolysis: ethanol induction of cytochrome P450 may potentiate myotoxicity. Alcohols 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 9844795-0 1998 Alcohol-associated rhabdomyolysis: ethanol induction of cytochrome P450 may potentiate myotoxicity. Ethanol 35-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 9844795-2 1998 Ethanol is a potent inducer of cytochrome P450. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 9844795-4 1998 Accordingly, some alcohol-associated myotoxicity could be related to skeletal muscle cytochrome P450 induction by ethanol leading to the production of toxic metabolites of other compounds that then injure muscle. Alcohols 18-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 9844795-4 1998 Accordingly, some alcohol-associated myotoxicity could be related to skeletal muscle cytochrome P450 induction by ethanol leading to the production of toxic metabolites of other compounds that then injure muscle. Ethanol 114-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 9844795-5 1998 The recent identification and localization of cytochrome P450 on skeletal muscle sarcoplasmic reticulum provides supportive evidence for this potential role of ethanol in the pathogenesis of alcohol-associated rhabdomyolysis. Ethanol 160-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 9844795-5 1998 The recent identification and localization of cytochrome P450 on skeletal muscle sarcoplasmic reticulum provides supportive evidence for this potential role of ethanol in the pathogenesis of alcohol-associated rhabdomyolysis. Alcohols 191-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 9840430-0 1998 Elucidation of individual cytochrome P450 enzymes involved in the metabolism of clozapine. Clozapine 80-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-41 9920346-0 1998 The cytochrome P450 suicide inhibitor, 1-aminobenzotriazole, sensitizes rats to zymosan-induced toxicity. 1-aminobenzotriazole 39-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 9920346-0 1998 The cytochrome P450 suicide inhibitor, 1-aminobenzotriazole, sensitizes rats to zymosan-induced toxicity. Zymosan 80-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 9825831-2 1998 To determine which cytochrome P450 is involved in taxotere biotransformation, 11 cDNA-expressed human cytochrome P450s were examined for their activity in the metabolism of taxotere and its derivatives. Docetaxel 50-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-117 9825831-2 1998 To determine which cytochrome P450 is involved in taxotere biotransformation, 11 cDNA-expressed human cytochrome P450s were examined for their activity in the metabolism of taxotere and its derivatives. Docetaxel 173-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 9825831-2 1998 To determine which cytochrome P450 is involved in taxotere biotransformation, 11 cDNA-expressed human cytochrome P450s were examined for their activity in the metabolism of taxotere and its derivatives. Docetaxel 173-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-117 9756035-2 1998 The present study investigated the role of human cytochrome P-450 (CYP) enzymes in sulfoxidation and thiono-oxidation of DDTC-Me, intermediary steps in the activation of disulfiram. methyl diethyldithiocarbamate 121-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 9756035-2 1998 The present study investigated the role of human cytochrome P-450 (CYP) enzymes in sulfoxidation and thiono-oxidation of DDTC-Me, intermediary steps in the activation of disulfiram. methyl diethyldithiocarbamate 121-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 9756035-2 1998 The present study investigated the role of human cytochrome P-450 (CYP) enzymes in sulfoxidation and thiono-oxidation of DDTC-Me, intermediary steps in the activation of disulfiram. Disulfiram 170-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 9756035-2 1998 The present study investigated the role of human cytochrome P-450 (CYP) enzymes in sulfoxidation and thiono-oxidation of DDTC-Me, intermediary steps in the activation of disulfiram. Disulfiram 170-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 9733665-0 1998 Human cytochrome P450-catalyzed conversion of the proestrogenic pesticide methoxychlor into an estrogen. Methoxychlor 74-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 9733665-3 1998 We have elucidated the human cytochrome P450 enzymes responsible for conversion of methoxychlor into its major metabolite, the mono-O-demethylated derivative (mono-OH-M) that is estrogenic. Methoxychlor 83-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 9733665-3 1998 We have elucidated the human cytochrome P450 enzymes responsible for conversion of methoxychlor into its major metabolite, the mono-O-demethylated derivative (mono-OH-M) that is estrogenic. mono-o 127-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 9733665-3 1998 We have elucidated the human cytochrome P450 enzymes responsible for conversion of methoxychlor into its major metabolite, the mono-O-demethylated derivative (mono-OH-M) that is estrogenic. mono-oh-m 159-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 9732390-0 1998 Application of liquid chromatography/mass spectrometry in accelerating the identification of human liver cytochrome P450 isoforms involved in the metabolism of iloperidone. iloperidone 160-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-120 9817620-2 1998 Cytochrome P450 (CYP) is a group of more than 30 different heme containing proteins in humans, some of which play a key role in the oxidation and hence the elimination of numerous drugs, including the SSRIs. Heme 59-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9817620-2 1998 Cytochrome P450 (CYP) is a group of more than 30 different heme containing proteins in humans, some of which play a key role in the oxidation and hence the elimination of numerous drugs, including the SSRIs. Heme 59-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 9732391-1 1998 The metabolism of etoposide was investigated by using human liver microsomes and nine recombinant human cytochrome P450 (CYP) isoforms to identify the CYP isoform(s) involved in the major metabolic pathway (3"-demethylation) of etoposide as well as to evaluate the possible metabolic interactions with several antitumor or supporting agents. Etoposide 228-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 9732391-3 1998 The relationships were assessed with six different human liver microsomes between the 3"-demethylation of etoposide and metabolic activities for substrate probes of the respective CYP isoforms, showing a significant correlation (r = 0. Etoposide 106-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 9732390-2 1998 The metabolism of iloperidone was studied in human liver microsomes to define the metabolic pathways and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of major iloperidone metabolites. iloperidone 18-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-136 9732390-2 1998 The metabolism of iloperidone was studied in human liver microsomes to define the metabolic pathways and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of major iloperidone metabolites. iloperidone 18-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 9732390-2 1998 The metabolism of iloperidone was studied in human liver microsomes to define the metabolic pathways and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of major iloperidone metabolites. iloperidone 191-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-136 9732391-7 1998 Among the nine recombinant human CYP isoforms, CYP3A4 exhibited the highest catalytic activity with respect to etoposide 3"-demethylation, compared with the minor activities of CYP1A2 and 2E1. etoposide 3" 111-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-36 9732390-2 1998 The metabolism of iloperidone was studied in human liver microsomes to define the metabolic pathways and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of major iloperidone metabolites. iloperidone 191-202 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 138-141 9732390-13 1998 The power of liquid chromatography/mass spectrometry in greatly accelerating the process of identifying the human liver CYP isoforms involved in the metabolism of iloperidone was demonstrated in this study. iloperidone 163-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 9732391-1 1998 The metabolism of etoposide was investigated by using human liver microsomes and nine recombinant human cytochrome P450 (CYP) isoforms to identify the CYP isoform(s) involved in the major metabolic pathway (3"-demethylation) of etoposide as well as to evaluate the possible metabolic interactions with several antitumor or supporting agents. Etoposide 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 9732391-1 1998 The metabolism of etoposide was investigated by using human liver microsomes and nine recombinant human cytochrome P450 (CYP) isoforms to identify the CYP isoform(s) involved in the major metabolic pathway (3"-demethylation) of etoposide as well as to evaluate the possible metabolic interactions with several antitumor or supporting agents. Etoposide 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 9732391-1 1998 The metabolism of etoposide was investigated by using human liver microsomes and nine recombinant human cytochrome P450 (CYP) isoforms to identify the CYP isoform(s) involved in the major metabolic pathway (3"-demethylation) of etoposide as well as to evaluate the possible metabolic interactions with several antitumor or supporting agents. Etoposide 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-154 10375803-1 1998 AIM: To study the effect of cytochrome P-450 (CYP450) inhibitors on clomipramine (Clo) N-demethylation in vitro. Clomipramine 68-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 9916257-0 1998 [Effect of cytochrome P450-dependent metabolites of arachidonic acid on the functional state of vessels]. Arachidonic Acid 52-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-26 9916257-1 1998 The present paper reviews recent studies about the role of the cytochrome P-450-dependent arachidonic acid metabolites. Arachidonic Acid 90-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 10375803-1 1998 AIM: To study the effect of cytochrome P-450 (CYP450) inhibitors on clomipramine (Clo) N-demethylation in vitro. Clomipramine 68-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-52 10375803-1 1998 AIM: To study the effect of cytochrome P-450 (CYP450) inhibitors on clomipramine (Clo) N-demethylation in vitro. Clomipramine 82-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 10375803-1 1998 AIM: To study the effect of cytochrome P-450 (CYP450) inhibitors on clomipramine (Clo) N-demethylation in vitro. Clomipramine 82-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-52 9767182-3 1998 Active oxygen species formed by cytochrome P-450 isoforms can simultaneously act as stimulators of proliferation and inhibitors of intercellular communications. Oxygen 7-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 9731223-3 1998 In this study, we investigated the formation of dopamine from trace amines, using human hepatic microsomes and human cytochrome P450 (CYP) isoforms expressed in yeast. Dopamine 48-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-132 9731223-3 1998 In this study, we investigated the formation of dopamine from trace amines, using human hepatic microsomes and human cytochrome P450 (CYP) isoforms expressed in yeast. Dopamine 48-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-137 9731223-4 1998 Among the 11 isoforms of human CYP expressed in yeast, CYP2D6 was the only isoform exhibiting strong ability to convert p-tyramine and m-tyramine to dopamine. Tyramine 120-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 9731223-4 1998 Among the 11 isoforms of human CYP expressed in yeast, CYP2D6 was the only isoform exhibiting strong ability to convert p-tyramine and m-tyramine to dopamine. 3-tyramine 135-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 9731223-4 1998 Among the 11 isoforms of human CYP expressed in yeast, CYP2D6 was the only isoform exhibiting strong ability to convert p-tyramine and m-tyramine to dopamine. Dopamine 149-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-34 9723818-9 1998 The changes in ethinyl oestradiol pharmacokinetics were consistent with an increase in clearance from enzymatic induction of glucuronidation and/or cytochrome P450 hydroxylation. Ethinyl Estradiol 15-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-163 9705755-1 1998 The dietary mutagens 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx) and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) are activated to genotoxins by rat and human liver cytochrome P450 (P450) 1A1- and 1A2-mediated N-oxidation. 2-amino-3,8-dimethylimidazo(4,5-f)quinoxaline 21-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-211 9705755-1 1998 The dietary mutagens 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx) and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) are activated to genotoxins by rat and human liver cytochrome P450 (P450) 1A1- and 1A2-mediated N-oxidation. 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine 79-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-211 9694927-2 1998 Both compounds use the same cytochrome-P450 metabolic pathway, resulting in an increase in plasma concentration of terfenadine. Terfenadine 115-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 9694927-12 1998 These potential dual actions on HERG currents suggest that precautions should be taken in long-term ketoconazole treatment, particularly for patients who have decreased liver function or are on a drug regimen requiring simultaneous medications that use cytochrome-P450 for breakdown, such as terfenadine or erythromycin, or Class III antiarrhythmic drugs. Terfenadine 292-303 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 253-268 9694927-12 1998 These potential dual actions on HERG currents suggest that precautions should be taken in long-term ketoconazole treatment, particularly for patients who have decreased liver function or are on a drug regimen requiring simultaneous medications that use cytochrome-P450 for breakdown, such as terfenadine or erythromycin, or Class III antiarrhythmic drugs. Ketoconazole 100-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 253-268 9701998-0 1998 [Methods of study on cytochrome P450 in relation to the metabolism and toxicity of alcohol and drugs]. Alcohols 83-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 9714539-2 1998 Here we report on the immobilisation of a fusion protein between plant cytochrome P450-71B1 (CYP71B1) and its electron donor, plant NADPH cytochrome P450 reductase using an oil-in-water macro-emulsion, termed polyaphron, which contains a proportion of internal organic phase (phi) greater than 0.74. Oils 173-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 9714539-2 1998 Here we report on the immobilisation of a fusion protein between plant cytochrome P450-71B1 (CYP71B1) and its electron donor, plant NADPH cytochrome P450 reductase using an oil-in-water macro-emulsion, termed polyaphron, which contains a proportion of internal organic phase (phi) greater than 0.74. Water 180-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 9714539-2 1998 Here we report on the immobilisation of a fusion protein between plant cytochrome P450-71B1 (CYP71B1) and its electron donor, plant NADPH cytochrome P450 reductase using an oil-in-water macro-emulsion, termed polyaphron, which contains a proportion of internal organic phase (phi) greater than 0.74. polyaphron 209-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 9651188-1 1998 We previously reported that 20-hydroxyeicosatetraenoic acid (20-HETE) is an endogenous cytochrome P450 (cP450) 4A metabolite of arachidonic acid (AA) in human lung tissue, and is a potent cyclooxygenase-dependent vasodilator of isolated pulmonary arteries. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 28-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 9651188-1 1998 We previously reported that 20-hydroxyeicosatetraenoic acid (20-HETE) is an endogenous cytochrome P450 (cP450) 4A metabolite of arachidonic acid (AA) in human lung tissue, and is a potent cyclooxygenase-dependent vasodilator of isolated pulmonary arteries. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 28-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-109 9651188-1 1998 We previously reported that 20-hydroxyeicosatetraenoic acid (20-HETE) is an endogenous cytochrome P450 (cP450) 4A metabolite of arachidonic acid (AA) in human lung tissue, and is a potent cyclooxygenase-dependent vasodilator of isolated pulmonary arteries. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 61-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 9651188-1 1998 We previously reported that 20-hydroxyeicosatetraenoic acid (20-HETE) is an endogenous cytochrome P450 (cP450) 4A metabolite of arachidonic acid (AA) in human lung tissue, and is a potent cyclooxygenase-dependent vasodilator of isolated pulmonary arteries. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 61-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-109 9651188-1 1998 We previously reported that 20-hydroxyeicosatetraenoic acid (20-HETE) is an endogenous cytochrome P450 (cP450) 4A metabolite of arachidonic acid (AA) in human lung tissue, and is a potent cyclooxygenase-dependent vasodilator of isolated pulmonary arteries. Arachidonic Acid 128-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 9651188-1 1998 We previously reported that 20-hydroxyeicosatetraenoic acid (20-HETE) is an endogenous cytochrome P450 (cP450) 4A metabolite of arachidonic acid (AA) in human lung tissue, and is a potent cyclooxygenase-dependent vasodilator of isolated pulmonary arteries. Arachidonic Acid 128-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-109 9721336-5 1998 It has been known for years that NO also binds to the heme moiety of cytochrome P-450 (CYP) with high affinity. Heme 54-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 9721336-5 1998 It has been known for years that NO also binds to the heme moiety of cytochrome P-450 (CYP) with high affinity. Heme 54-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 9721340-6 1998 The reduction of NO2- ions to NO is realized by electron-donor systems with the participation of NADH, NADPH, flavoproteins, and cytochrome oxidase in mitochondria and by NADH, NADPH, flavoproteins, and cytochrome P-450 in endoplasmic reticulum. Nitrogen Dioxide 17-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 203-219 9616188-1 1998 Classic antihistamines, namely diphenhydramine, chlorpheniramine, clemastine, perphenazine, hydroxyzine, and tripelennamine, share structural features with substrates and inhibitors of the polymorphic cytochrome P450 (CYP) isozyme CYP2D6. Diphenhydramine 31-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-216 9616188-1 1998 Classic antihistamines, namely diphenhydramine, chlorpheniramine, clemastine, perphenazine, hydroxyzine, and tripelennamine, share structural features with substrates and inhibitors of the polymorphic cytochrome P450 (CYP) isozyme CYP2D6. Chlorpheniramine 48-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-216 9616188-1 1998 Classic antihistamines, namely diphenhydramine, chlorpheniramine, clemastine, perphenazine, hydroxyzine, and tripelennamine, share structural features with substrates and inhibitors of the polymorphic cytochrome P450 (CYP) isozyme CYP2D6. Clemastine 66-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-216 9616188-1 1998 Classic antihistamines, namely diphenhydramine, chlorpheniramine, clemastine, perphenazine, hydroxyzine, and tripelennamine, share structural features with substrates and inhibitors of the polymorphic cytochrome P450 (CYP) isozyme CYP2D6. Perphenazine 78-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-216 9616188-1 1998 Classic antihistamines, namely diphenhydramine, chlorpheniramine, clemastine, perphenazine, hydroxyzine, and tripelennamine, share structural features with substrates and inhibitors of the polymorphic cytochrome P450 (CYP) isozyme CYP2D6. Hydroxyzine 92-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-216 9616188-1 1998 Classic antihistamines, namely diphenhydramine, chlorpheniramine, clemastine, perphenazine, hydroxyzine, and tripelennamine, share structural features with substrates and inhibitors of the polymorphic cytochrome P450 (CYP) isozyme CYP2D6. Tripelennamine 109-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-216 9744552-0 1998 An evaluation of the cytochrome P450 induction potential of pantoprazole in primary human hepatocytes. Pantoprazole 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 9744552-2 1998 In this study, the cytochrome P450 (CYP) induction potential of pantoprazole (PAN) was evaluated and compared to two other proton pump inhibitors (PPIs), omeprazole (OM) and lansoprazole (LAN). Pantoprazole 64-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 9744552-2 1998 In this study, the cytochrome P450 (CYP) induction potential of pantoprazole (PAN) was evaluated and compared to two other proton pump inhibitors (PPIs), omeprazole (OM) and lansoprazole (LAN). Pantoprazole 64-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 9705898-8 1998 Experiments using human cell line cells either expressing or not expressing human cytochrome P450 enzymes in assays of acute toxicity (MTT assays) have demonstrated a directly toxic potential of AFM1 in the absence of metabolic activation, in contrast to AFB1. monooxyethylene trimethylolpropane tristearate 135-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 9771309-0 1998 Possible involvement of multiple human cytochrome P450 isoforms in the liver metabolism of propofol. Propofol 91-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 9771309-2 1998 On the basis of potential influence of propofol on the metabolism of co-administered agents, many investigators have evaluated the effects of propofol on cytochrome P450 (CYP) activities. Propofol 142-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 9771309-2 1998 On the basis of potential influence of propofol on the metabolism of co-administered agents, many investigators have evaluated the effects of propofol on cytochrome P450 (CYP) activities. Propofol 142-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-174 9771309-3 1998 CYP isoforms involved in propofol metabolism are not defined. Propofol 25-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 9771309-4 1998 In this study, our objective was to elucidate further the CYP isoforms responsible for the hydroxylation of propofol. Propofol 108-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-61 9771309-10 1998 This low specificity among CYP isoforms may contribute to the low interindividual variability (two-fold) and may contribute to the low level of metabolic drug interactions observed with propofol. Propofol 186-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 9616191-3 1998 Ritonavir was similarly biotransformed by microsomes containing expressed CYP3A4 or CYP3A5 isozymes (KM = 0.05-0.07 microM, Vmax = 1-1.4 nmol/min/nmol CYP). Ritonavir 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 9616191-4 1998 In contrast, both the patterns of metabolites and the enzyme kinetic parameters for the metabolism of indinavir by expressed CYP3A5 (KM = 0.21 microM, Vmax = 0.24 nmol/min/nmol CYP) and CYP3A4 (KM = 0.04 microM, Vmax = 0.68 nmol/min/nmol CYP) were different. Indinavir 102-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-128 9616191-4 1998 In contrast, both the patterns of metabolites and the enzyme kinetic parameters for the metabolism of indinavir by expressed CYP3A5 (KM = 0.21 microM, Vmax = 0.24 nmol/min/nmol CYP) and CYP3A4 (KM = 0.04 microM, Vmax = 0.68 nmol/min/nmol CYP) were different. Indinavir 102-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-180 9616193-0 1998 N-Dealkylation and hydroxylation of ebastine by human liver cytochrome P450. ebastine 36-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 9616193-2 1998 The present study was performed to characterize the cytochrome P450 (CYP) isoforms responsible for ebastine N-dealkylation and hydroxylation. ebastine 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 9616193-2 1998 The present study was performed to characterize the cytochrome P450 (CYP) isoforms responsible for ebastine N-dealkylation and hydroxylation. ebastine 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 9616193-5 1998 In a panel of 14 human liver microsomal preparations, the rate of dealkylation showed a highly significant correlation with CYP3A-mediated testosterone 6beta-hydroxylation but not with reactions of seven other CYP isoforms. Testosterone 139-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-127 9616193-9 1998 Among 12 cDNA-expressed human CYP isoforms, which account for up to 70% of the total CYP enzyme content in human liver, CYP3A4 alone metabolized ebastine; the ratio of des-BP to M-OH formation was 12:1. ebastine 145-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 9578592-1 1998 In this paper two hypotheses are tested: (i) the active oxygen species is similar in energetics for all cytochrome P450 (CYP) enzymes and (ii) linear free-energy relationships can be used to evaluate the mechanism of the reaction of these enzymes. Oxygen 56-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-119 9578592-0 1998 Evaluation of cytochrome P450 mechanism and kinetics using kinetic deuterium isotope effects. Deuterium 67-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 9578592-1 1998 In this paper two hypotheses are tested: (i) the active oxygen species is similar in energetics for all cytochrome P450 (CYP) enzymes and (ii) linear free-energy relationships can be used to evaluate the mechanism of the reaction of these enzymes. Oxygen 56-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 9606477-7 1998 Phenytoin, phenobarbital, and carbamazepine induce CYP and UGT enzymes. Phenytoin 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 9606477-7 1998 Phenytoin, phenobarbital, and carbamazepine induce CYP and UGT enzymes. Phenobarbital 11-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 9606477-7 1998 Phenytoin, phenobarbital, and carbamazepine induce CYP and UGT enzymes. Carbamazepine 30-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-54 9635877-2 1998 They are thought to be metabolically activated by N-hydroxylation, catalysed by cytochrome P450 (CYP), followed by O-acetylation catalysed by N-acetyltransferases. Nitrogen 50-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 9635877-2 1998 They are thought to be metabolically activated by N-hydroxylation, catalysed by cytochrome P450 (CYP), followed by O-acetylation catalysed by N-acetyltransferases. Nitrogen 50-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 9635877-7 1998 The higher adduct levels observed in cells treated with the N-OH metabolites suggests that N-hydroxylation is the rate-limiting step in HMECs and this may be due to low CYP levels. n-oh 60-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 9635877-7 1998 The higher adduct levels observed in cells treated with the N-OH metabolites suggests that N-hydroxylation is the rate-limiting step in HMECs and this may be due to low CYP levels. Nitrogen 60-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-172 9594936-11 1998 Itraconazole has an affinity for mammalian cytochrome P-450 enzymes as well as for fungal P-450-dependent enzyme, and thus has the potential for clinically important interactions (e.g., astemizole, terfenadine, rifampin, oral contraceptives, H2 receptor antagonists, warfarin, cyclosporine). Itraconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 9630823-1 1998 OBJECTIVE: To identify the cytochrome P450 (CYP) enzymes involved in the conversion of vesnarinone to it main primary metabolite OPC-18692 and to investigate the effect of CYP3A inhibition on the pharmacokinetics of vesnarinone in vivo. vesnarinone 87-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 9630823-1 1998 OBJECTIVE: To identify the cytochrome P450 (CYP) enzymes involved in the conversion of vesnarinone to it main primary metabolite OPC-18692 and to investigate the effect of CYP3A inhibition on the pharmacokinetics of vesnarinone in vivo. vesnarinone 87-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 9630823-1 1998 OBJECTIVE: To identify the cytochrome P450 (CYP) enzymes involved in the conversion of vesnarinone to it main primary metabolite OPC-18692 and to investigate the effect of CYP3A inhibition on the pharmacokinetics of vesnarinone in vivo. vesnarinone 216-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 9630823-1 1998 OBJECTIVE: To identify the cytochrome P450 (CYP) enzymes involved in the conversion of vesnarinone to it main primary metabolite OPC-18692 and to investigate the effect of CYP3A inhibition on the pharmacokinetics of vesnarinone in vivo. vesnarinone 216-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 9654063-4 1998 A reaction pathway like that of cytochrome P-450 implies oxygen transfer to the substrate from the as yet uncharacterized iron-oxo species formed in the reaction of the heine cofactor with H2O2. Oxygen 57-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 9654063-4 1998 A reaction pathway like that of cytochrome P-450 implies oxygen transfer to the substrate from the as yet uncharacterized iron-oxo species formed in the reaction of the heine cofactor with H2O2. iron-oxo 122-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 9654063-4 1998 A reaction pathway like that of cytochrome P-450 implies oxygen transfer to the substrate from the as yet uncharacterized iron-oxo species formed in the reaction of the heine cofactor with H2O2. Hydrogen Peroxide 189-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 9654063-6 1998 The results of the present study exclude Fenton-type chemistry and prove that the minicatalyst is able to catalyze the oxygen incorporation by both peroxidase and cytochrome P-450 types of reaction pathways, while exchange occurs between the high-valency iron-oxo species and H2O. Oxygen 119-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 9654063-6 1998 The results of the present study exclude Fenton-type chemistry and prove that the minicatalyst is able to catalyze the oxygen incorporation by both peroxidase and cytochrome P-450 types of reaction pathways, while exchange occurs between the high-valency iron-oxo species and H2O. iron-oxo 255-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 9654063-6 1998 The results of the present study exclude Fenton-type chemistry and prove that the minicatalyst is able to catalyze the oxygen incorporation by both peroxidase and cytochrome P-450 types of reaction pathways, while exchange occurs between the high-valency iron-oxo species and H2O. Water 276-279 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 9580580-0 1998 Identification and characterization of human cytochrome P450 isoforms interacting with pimozide. Pimozide 87-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-60 9580580-1 1998 Using human liver microsomes (HLMs) and recombinant human cytochrome P450 (CYP450) isoforms, we identified the major route of pimozide metabolism, the CYP450 isoforms involved, and documented the inhibitory effect of pimozide on CYP450 isoforms. Pimozide 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 9518732-23 1998 The present data suggest that cytochrome P450 modulates cardiac ICa and cell contraction, and the modulation may result from changes in intracellular levels of cAMP by P450- mediated metabolites of arachidonic acid. Cyclic AMP 160-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 9518732-23 1998 The present data suggest that cytochrome P450 modulates cardiac ICa and cell contraction, and the modulation may result from changes in intracellular levels of cAMP by P450- mediated metabolites of arachidonic acid. Arachidonic Acid 198-214 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 9580580-1 1998 Using human liver microsomes (HLMs) and recombinant human cytochrome P450 (CYP450) isoforms, we identified the major route of pimozide metabolism, the CYP450 isoforms involved, and documented the inhibitory effect of pimozide on CYP450 isoforms. Pimozide 126-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-81 9591923-3 1998 Ethanol is consumed worldwide in tremendous amounts and is an effective inducer of hepatic drug metabolism, especially involving pathways accomplished by the CYP2E1 isoform of the cytochrome P-450 (CYP) superfamily. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 9579827-3 1998 Liarozole-fumarate is an anti-tumour drug that inhibits the cytochrome P450-dependent catabolism of ATRA. liarozole 0-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 9579827-3 1998 Liarozole-fumarate is an anti-tumour drug that inhibits the cytochrome P450-dependent catabolism of ATRA. Tretinoin 100-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 9531517-0 1998 Metabolism of beta-arteether to dihydroqinghaosu by human liver microsomes and recombinant cytochrome P450. artemotil 14-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-106 9531517-2 1998 Studies were undertaken to determine which form(s) of human cytochrome P-450 catalyze the conversion of beta-arteether to its deethylated metabolite, dihydroqinghaosu (DQHS), itself a potent antimalarial compound. artemotil 104-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 9531517-2 1998 Studies were undertaken to determine which form(s) of human cytochrome P-450 catalyze the conversion of beta-arteether to its deethylated metabolite, dihydroqinghaosu (DQHS), itself a potent antimalarial compound. artenimol 150-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 9556346-0 1998 Step-scan time-resolved FTIR spectroscopy of cytochrome P-450cam carbon monoxide complex: a salt link involved in the ligand-rebinding process. Carbon Monoxide 65-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 9556346-0 1998 Step-scan time-resolved FTIR spectroscopy of cytochrome P-450cam carbon monoxide complex: a salt link involved in the ligand-rebinding process. Salts 92-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 9539102-1 1998 Leukotriene B4 (LTB4) omega-hydroxylase catalyzes the conversion of LTB4 into a biologically less active product, 20-hydroxy-LTB4. 5,12,20-trihydroxy-6,8,10,14-eicosatetraenoic acid 114-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-39 9539102-5 1998 The cDNA of CYP4F3 contained 5050 nucleotides excluding the poly(A) tail. Poly A 60-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-18 9533981-9 1998 Higher plasma drug concentrations may also be obtained by combining the drug with CYP blockers, such as ritonavir or ketoconazole. Ritonavir 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 9533981-10 1998 Because of its metabolic interference with the CYP system, saquinavir cannot be coadministered with astemizole, terfenadine or cisapride. Saquinavir 59-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-50 9492390-2 1998 We studied the human cytochrome P450 (CYP) isoform responsible for indomethacin O-demethylation, the major metabolic pathway for indomethacin. Indomethacin 67-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 9492390-2 1998 We studied the human cytochrome P450 (CYP) isoform responsible for indomethacin O-demethylation, the major metabolic pathway for indomethacin. Indomethacin 67-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 9492390-2 1998 We studied the human cytochrome P450 (CYP) isoform responsible for indomethacin O-demethylation, the major metabolic pathway for indomethacin. Indomethacin 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 9591923-3 1998 Ethanol is consumed worldwide in tremendous amounts and is an effective inducer of hepatic drug metabolism, especially involving pathways accomplished by the CYP2E1 isoform of the cytochrome P-450 (CYP) superfamily. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-161 9492390-2 1998 We studied the human cytochrome P450 (CYP) isoform responsible for indomethacin O-demethylation, the major metabolic pathway for indomethacin. Indomethacin 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 9600717-6 1998 Ketoconazole, a relatively specific CYP 3A4 inhibitor, inhibited the reaction; the concentration resulting in 50% of maximum inhibition, IC50, was 0.15 microM and the inhibition constant, Ki, was < 0.04 microM in five of six livers tested. Ketoconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 9523980-5 1998 An in vitro metabolic study using recombinant human cytochrome P450 (CYP) isoforms indicated that oxidation of both bisoprolol enantiomers was catalyzed by the two isoforms, CYP2D6 and CYP3A4. Bisoprolol 116-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 9523980-5 1998 An in vitro metabolic study using recombinant human cytochrome P450 (CYP) isoforms indicated that oxidation of both bisoprolol enantiomers was catalyzed by the two isoforms, CYP2D6 and CYP3A4. Bisoprolol 116-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-72 9600717-12 1998 These results show that CYP 3A4 is the primary hepatic CYP isoform mediating the N-demethylation of adinazolam and NDMAD. Nitrogen 81-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 9600717-12 1998 These results show that CYP 3A4 is the primary hepatic CYP isoform mediating the N-demethylation of adinazolam and NDMAD. adinazolam 100-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 9600717-12 1998 These results show that CYP 3A4 is the primary hepatic CYP isoform mediating the N-demethylation of adinazolam and NDMAD. mono-N-demethyladinazolam 115-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 9600717-13 1998 Co-administration of adinazolam with CYP 3A4 inhibitors such as ketoconazole or erythromycin might lead to reduced efficacy, since adinazolam by itself has relatively weak benzodiazepine agonist activity, with much of the pharmacological activity of adinazolam being attributable to its active metabolite NDMAD. Ketoconazole 64-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 9600717-13 1998 Co-administration of adinazolam with CYP 3A4 inhibitors such as ketoconazole or erythromycin might lead to reduced efficacy, since adinazolam by itself has relatively weak benzodiazepine agonist activity, with much of the pharmacological activity of adinazolam being attributable to its active metabolite NDMAD. Erythromycin 80-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 9600717-13 1998 Co-administration of adinazolam with CYP 3A4 inhibitors such as ketoconazole or erythromycin might lead to reduced efficacy, since adinazolam by itself has relatively weak benzodiazepine agonist activity, with much of the pharmacological activity of adinazolam being attributable to its active metabolite NDMAD. adinazolam 131-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 9600717-13 1998 Co-administration of adinazolam with CYP 3A4 inhibitors such as ketoconazole or erythromycin might lead to reduced efficacy, since adinazolam by itself has relatively weak benzodiazepine agonist activity, with much of the pharmacological activity of adinazolam being attributable to its active metabolite NDMAD. adinazolam 131-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-40 9442307-6 1998 At the biochemical level the tests can indicate that the organism has been exposed to certain groups of chemicals (for example cytochrome P-450 enzymes responding to PAHs or metallothioneins responding to heavy metals). Polycyclic Aromatic Hydrocarbons 166-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-143 9481028-2 1998 Premature infants have reduced activity of cytochrome p-450, the system responsible for metabolism of cisapride, which could lead to QT prolongation. Cisapride 102-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 9454825-7 1998 The observed changes in the activity and protein levels of CYP1A2 and CYP3A4 correlated with a reduction in the specific messenger RNA levels both in control, 3-methylcholanthrene-treated (for CYP1A2) and rifampicin-treated (for CYP3A4) hepatocytes, which thus suggested that HGF could down-regulate CYP expression at a pretranslational level. Methylcholanthrene 159-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 9454825-7 1998 The observed changes in the activity and protein levels of CYP1A2 and CYP3A4 correlated with a reduction in the specific messenger RNA levels both in control, 3-methylcholanthrene-treated (for CYP1A2) and rifampicin-treated (for CYP3A4) hepatocytes, which thus suggested that HGF could down-regulate CYP expression at a pretranslational level. Rifampin 205-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 9458080-1 1998 Isothiocyanates exert strong anticarcinogenic effects in a number of animal models of cancer, presumably by modulation of xenobiotic-metabolizing enzymes, such as by inhibition of cytochrome P-450 and/or by induction of phase II detoxifying enzymes. Isothiocyanates 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 9498238-0 1998 Expression of cytochrome P450 genes encoding enzymes active in the metabolism of tamoxifen in human uterine endometrium. Tamoxifen 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-29 9498238-7 1998 These data suggest that the human endometrial epithelium has the potential of producing CYP enzymes known to generate genotoxic intermediates from tamoxifen and metabolites that affect oestrogen receptors. Tamoxifen 147-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 9493761-17 1998 Examples of human teratogens that are substrates for CYP enzymes include thalidomide, phenytoin, ethanol, and several hormonal agents. Thalidomide 73-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 9493761-17 1998 Examples of human teratogens that are substrates for CYP enzymes include thalidomide, phenytoin, ethanol, and several hormonal agents. Phenytoin 86-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 9493761-17 1998 Examples of human teratogens that are substrates for CYP enzymes include thalidomide, phenytoin, ethanol, and several hormonal agents. Ethanol 97-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 9763205-5 1998 The inhibition studies in dog liver microsomes using CYP isoform-selective inhibitors indicated that the O-deisopropylation of both bisoprolol enantiomers was mediated via the CYP2D and CYP3A subfamily, and suggested that high-affinity oxidation was dependent on CYP2D. Bisoprolol 132-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 9740537-7 1998 Unlike other drugs of its class, cerivastatin has a dual metabolic pathway, with the involvement of more than one CYP isozyme. cerivastatin 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 9543613-2 1998 Therefore, drugs such as cimetidine, which inhibit the cytochrome P-450 enzyme system or reduce liver blood flow, may reduce the plasma clearance of epirubicin. Cimetidine 25-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 9543613-2 1998 Therefore, drugs such as cimetidine, which inhibit the cytochrome P-450 enzyme system or reduce liver blood flow, may reduce the plasma clearance of epirubicin. Epirubicin 149-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 9763205-6 1998 The kinds of CYP subfamilies in dogs, which contribute to the metabolism of bisoprolol enantiomers, were the same as those in humans. Bisoprolol 76-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-16 9763205-9 1998 Therefore, the species difference in the R/S ratio of metabolic clearance for the oxidation of bisoprolol enantiomers (dog > human) is mainly due to the species difference in the stereoselectivity of one of the cytochrome P450 subfamilies (CYP3A). Bisoprolol 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 214-229 9394028-13 1997 The correlation data was confirmed by inhibition studies and cDNA expressed cytochrome P450 systems demonstrating that the biotransformation of tazofelone to its metabolites is primarily mediated by CYP3A. tazofelone 144-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 9366245-7 1997 This data suggests that metabolism of hepoxilin A3 occurs in intact human neutrophils through omega-oxidation, is likely located in the mitochondrial compartment of the cell (inhibition by CCCP) and is carried out by an activity that is independent of the well characterized, relatively stable microsomal LTB4 omega-hydroxylase. 8-hydroxy-11,12-epoxyeicosa-5,9,14-trienoic acid 38-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 305-327 9394024-0 1997 Cytochrome P450 isozymes involved in lisofylline metabolism to pentoxifylline in human liver microsomes. lisofylline 37-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9394024-0 1997 Cytochrome P450 isozymes involved in lisofylline metabolism to pentoxifylline in human liver microsomes. Pentoxifylline 63-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9394024-1 1997 We describe the kinetics of pentoxifylline formation from lisofylline in human liver microsomes using selective inhibitors of cytochrome P450 isozymes, correlation studies with specific isozyme activities, and cDNA-expressed human CYP1A2 and 2E1. Pentoxifylline 28-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-141 9394024-9 1997 Baculovirus insect cell expressed human CYP1A2 formed pentoxifylline at 0.987 nmol/min/nmol cytochrome P450 at 5 microM lisofylline. Pentoxifylline 54-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 9394024-9 1997 Baculovirus insect cell expressed human CYP1A2 formed pentoxifylline at 0.987 nmol/min/nmol cytochrome P450 at 5 microM lisofylline. lisofylline 120-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 9394027-0 1997 Metabolism of clozapine by cDNA-expressed human cytochrome P450 enzymes. Clozapine 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 9394027-1 1997 The metabolism of clozapine was studied in vitro using cDNA-expressed human cytochrome P450 (CYP) enzymes 1A2, 3A4, 2C9, 2C19, 2D6, and 2E1. Clozapine 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-91 9394027-1 1997 The metabolism of clozapine was studied in vitro using cDNA-expressed human cytochrome P450 (CYP) enzymes 1A2, 3A4, 2C9, 2C19, 2D6, and 2E1. Clozapine 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-96 9394028-0 1997 In vitro biotransformation and identification of human cytochrome P450 isozyme-dependent metabolism of tazofelone. tazofelone 103-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-70 9394028-8 1997 Tazofelone was incubated at 20 microM concentration with human microsomes to determine which of the cytochrome P450 isozyme(s) is involved in the oxidation of tazofelone. tazofelone 159-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 9429083-6 1997 Quercetin inhibited activity of human cytochrome P-450 towards ethoxycoumarin and ethylresorufin at relatively low substrate concentrations (0.1 microM and above). Quercetin 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 9429083-6 1997 Quercetin inhibited activity of human cytochrome P-450 towards ethoxycoumarin and ethylresorufin at relatively low substrate concentrations (0.1 microM and above). 3-ethoxychromen-2-one 63-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 9429083-6 1997 Quercetin inhibited activity of human cytochrome P-450 towards ethoxycoumarin and ethylresorufin at relatively low substrate concentrations (0.1 microM and above). ethylresorufin 82-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 9385444-1 1997 Human cytochrome P450 (P450) 3A is known to be involved in the formation of both aflatoxin B1-exo-8,9-epoxide (exo-epoxidation) and aflatoxin Q1 (3 alpha-hydroxylation). aflatoxin B1-2,3-oxide 81-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-31 9385444-1 1997 Human cytochrome P450 (P450) 3A is known to be involved in the formation of both aflatoxin B1-exo-8,9-epoxide (exo-epoxidation) and aflatoxin Q1 (3 alpha-hydroxylation). aflatoxin Q1 132-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-31 9399333-0 1997 Metabolism of the tricyclic antidepressant amitriptyline by cDNA-expressed human cytochrome P450 enzymes. Amitriptyline 43-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 9399333-1 1997 The metabolism of amitriptyline was studied in vitro using cDNA-expressed human cytochrome P450 (CYP) enzymes 1A2, 3A4, 2C9, 2C19, 2D6 and 2E1. Amitriptyline 18-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 9399333-1 1997 The metabolism of amitriptyline was studied in vitro using cDNA-expressed human cytochrome P450 (CYP) enzymes 1A2, 3A4, 2C9, 2C19, 2D6 and 2E1. Amitriptyline 18-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 9385296-1 1997 Rifampin is a potent inducer of cytochrome P-450 oxidative enzymes. Rifampin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 9357977-0 1997 Crystal structure of cytochrome P-450cam complexed with the (1S)-camphor enantiomer. Camphor 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 9357977-1 1997 The crystal structure of cytochrome P-450cam complexed with the enantiomer (1S)-camphor has been solved to 1.8 angstroms resolution and compared with the structure of the (1R)-camphor P-450cam complex. Camphor 80-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 9357977-1 1997 The crystal structure of cytochrome P-450cam complexed with the enantiomer (1S)-camphor has been solved to 1.8 angstroms resolution and compared with the structure of the (1R)-camphor P-450cam complex. (1r)-camphor p 171-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 9353130-14 1997 These results demonstrate that rat and human hepatocytes preserve the major forms of CYP isozymes and phase II activities and respond to inducing drugs such as rifampicin. Rifampin 160-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 9350042-0 1997 A microassay for the detection of low levels of cytochrome P450 O-deethylation activities with alkoxyresorufin substrates. alkoxyresorufin 95-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 9353130-3 1997 Basal levels of cytochrome P-450 (CYP) isozymes were characterized by measuring ethoxyresorufin O-deethylation (EROD), ethoxycoumarin O-deethylation (ECOD), and the specific oxidation of testosterone (T). ethoxyresorufin 80-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 9353130-3 1997 Basal levels of cytochrome P-450 (CYP) isozymes were characterized by measuring ethoxyresorufin O-deethylation (EROD), ethoxycoumarin O-deethylation (ECOD), and the specific oxidation of testosterone (T). ethoxyresorufin 80-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 9353130-3 1997 Basal levels of cytochrome P-450 (CYP) isozymes were characterized by measuring ethoxyresorufin O-deethylation (EROD), ethoxycoumarin O-deethylation (ECOD), and the specific oxidation of testosterone (T). 3-ethoxychromen-2-one 119-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 9353130-3 1997 Basal levels of cytochrome P-450 (CYP) isozymes were characterized by measuring ethoxyresorufin O-deethylation (EROD), ethoxycoumarin O-deethylation (ECOD), and the specific oxidation of testosterone (T). Testosterone 187-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 9353130-3 1997 Basal levels of cytochrome P-450 (CYP) isozymes were characterized by measuring ethoxyresorufin O-deethylation (EROD), ethoxycoumarin O-deethylation (ECOD), and the specific oxidation of testosterone (T). Testosterone 187-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-37 9391787-0 1997 Relationship between cytochrome P-450 induction by rifampicin, hepatic volume and portal blood flow in man. Rifampin 51-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 9391787-10 1997 CONCLUSION: A fourfold increase of urinary 6-beta-hydroxycortisol output after induction of cytochrome P-450 by rifampicin is associated with a significant but less than 10% increase in human liver volume. 6 beta-hydroxycortisol 43-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 9391787-10 1997 CONCLUSION: A fourfold increase of urinary 6-beta-hydroxycortisol output after induction of cytochrome P-450 by rifampicin is associated with a significant but less than 10% increase in human liver volume. Rifampin 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 9358560-0 1997 Biomimetic metabolism of artelinic acid by chemical cytochrome P-450 model systems. artelinic acid 25-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 9430380-0 1997 Cytochrome P450 metabolites of arachidonic acid: rapid incorporation and hydration of 14,15-epoxyeicosatrienoic acid in arterial smooth muscle cells. Arachidonic Acid 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9430380-0 1997 Cytochrome P450 metabolites of arachidonic acid: rapid incorporation and hydration of 14,15-epoxyeicosatrienoic acid in arterial smooth muscle cells. 14,15-epoxy-5,8,11-eicosatrienoic acid 86-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9430380-1 1997 Arachidonic acid is converted to epoxyeicosatrienoic acids (EETs) by cytochrome P450 monooxygenases. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 9440131-0 1997 Cytochrome P450 metabolites of arachidonic acid in human placenta. Arachidonic Acid 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9358560-1 1997 PURPOSE: To study the reaction of artelinic acid with chemical model systems of cytochrome P-450 as a means of obtaining authentic samples of the putative metabolites necessary for identification of the mammalian metabolites of artelinic acid. artelinic acid 34-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 9430380-1 1997 Arachidonic acid is converted to epoxyeicosatrienoic acids (EETs) by cytochrome P450 monooxygenases. epoxyeicosatrienoic acids 33-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 9358560-1 1997 PURPOSE: To study the reaction of artelinic acid with chemical model systems of cytochrome P-450 as a means of obtaining authentic samples of the putative metabolites necessary for identification of the mammalian metabolites of artelinic acid. artelinic acid 228-242 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 9430380-1 1997 Arachidonic acid is converted to epoxyeicosatrienoic acids (EETs) by cytochrome P450 monooxygenases. eets 60-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 9358560-5 1997 CONCLUSIONS: Standards of possible metabolites of artelinic acid can be produced by the reaction of the compound with ferrous complexes that may simulate cytochrome P-450 catalyzed metabolism of xenobiotics. artelinic acid 50-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-170 9344892-0 1997 Metabolism of ethyl carbamate by pulmonary cytochrome P450 and carboxylesterase isozymes: involvement of CYP2E1 and hydrolase A. Urethane 14-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-79 9361171-1 1997 BACKGROUND: Omeprazole is eliminated almost completely by hepatic metabolism within the cytochrome P-450 system and might inhibit the oxidative metabolism of other drugs. Omeprazole 12-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 9344892-2 1997 Our goal in this study was to investigate the in vitro isozyme-selective metabolism of EC in lung microsomes by cytochrome P450 and carboxylesterase enzymes. Urethane 87-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-148 9303380-1 1997 Fluconazole, an inhibitor of certain human cytochrome P-450 isozymes, is used for the prevention and treatment of a broad range of fungal infections that predominantly affect immunocompromised individuals. Fluconazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 9526800-4 1997 The way to regulate ALAS-N in the liver is suitable to supply a constant level of heme for a family of drug metabolizing enzymes, cytochrome P-450 (CYP). Heme 82-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 9526800-4 1997 The way to regulate ALAS-N in the liver is suitable to supply a constant level of heme for a family of drug metabolizing enzymes, cytochrome P-450 (CYP). Heme 82-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-151 9526800-7 1997 ALAS-N in undifferentiated erythroid cells, therefore, is suggested to produce heme for CYP, whereas heme for accumulating hemoglobin (Hb) in cells undergoing differentiation is synthesized via ALAS-E. Heme 79-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-91 9270401-2 1997 TDP has been associated with terfenadine use in cases of liver disease, electrolyte abnormalities, concomitant administration of drugs that inhibit cytochrome P-450, or deliberate overdose. Terfenadine 29-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 9381730-0 1997 Examination of metabolic pathways and identification of human liver cytochrome P450 isozymes responsible for the metabolism of barnidipine, a calcium channel blocker. mepirodipine 127-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 9305589-0 1997 Bioactivation of phenytoin by human cytochrome P450: characterization of the mechanism and targets of covalent adduct formation. Phenytoin 17-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 9305589-1 1997 The cytochrome P450-dependent covalent binding of radiolabel derived from phenytoin (DPH) and its phenol and catechol metabolites, 5-(4"-hydroxyphenyl)-5-phenylhydantoin (HPPH) and 5-(3",4"-dihydroxyphenyl)-5-phenylhydantoin (CAT), was examined in liver microsomes. Phenytoin 74-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 9305589-1 1997 The cytochrome P450-dependent covalent binding of radiolabel derived from phenytoin (DPH) and its phenol and catechol metabolites, 5-(4"-hydroxyphenyl)-5-phenylhydantoin (HPPH) and 5-(3",4"-dihydroxyphenyl)-5-phenylhydantoin (CAT), was examined in liver microsomes. Phenytoin 85-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 9305589-1 1997 The cytochrome P450-dependent covalent binding of radiolabel derived from phenytoin (DPH) and its phenol and catechol metabolites, 5-(4"-hydroxyphenyl)-5-phenylhydantoin (HPPH) and 5-(3",4"-dihydroxyphenyl)-5-phenylhydantoin (CAT), was examined in liver microsomes. Phenol 98-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 9305589-1 1997 The cytochrome P450-dependent covalent binding of radiolabel derived from phenytoin (DPH) and its phenol and catechol metabolites, 5-(4"-hydroxyphenyl)-5-phenylhydantoin (HPPH) and 5-(3",4"-dihydroxyphenyl)-5-phenylhydantoin (CAT), was examined in liver microsomes. catechol 109-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 9305589-1 1997 The cytochrome P450-dependent covalent binding of radiolabel derived from phenytoin (DPH) and its phenol and catechol metabolites, 5-(4"-hydroxyphenyl)-5-phenylhydantoin (HPPH) and 5-(3",4"-dihydroxyphenyl)-5-phenylhydantoin (CAT), was examined in liver microsomes. hydroxyphenytoin 131-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 9305589-1 1997 The cytochrome P450-dependent covalent binding of radiolabel derived from phenytoin (DPH) and its phenol and catechol metabolites, 5-(4"-hydroxyphenyl)-5-phenylhydantoin (HPPH) and 5-(3",4"-dihydroxyphenyl)-5-phenylhydantoin (CAT), was examined in liver microsomes. hydroxyphenytoin 171-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 9305589-1 1997 The cytochrome P450-dependent covalent binding of radiolabel derived from phenytoin (DPH) and its phenol and catechol metabolites, 5-(4"-hydroxyphenyl)-5-phenylhydantoin (HPPH) and 5-(3",4"-dihydroxyphenyl)-5-phenylhydantoin (CAT), was examined in liver microsomes. 5-(3,4-dihydroxyphenyl)-5-phenylhydantoin 181-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 9316860-1 1997 Cytochrome P450 (CYP) and uridine diphosphate glucuronosyltransferase (UGT) isoenzymes involved in riluzole oxidation and glucuronidation were characterized in (1) kinetic studies with human hepatic microsomes and isoenzyme-selective probes and (2) metabolic studies with genetically expressed human CYP isoenzymes from transfected B-lymphoblastoid and yeast cells. Riluzole 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9316860-1 1997 Cytochrome P450 (CYP) and uridine diphosphate glucuronosyltransferase (UGT) isoenzymes involved in riluzole oxidation and glucuronidation were characterized in (1) kinetic studies with human hepatic microsomes and isoenzyme-selective probes and (2) metabolic studies with genetically expressed human CYP isoenzymes from transfected B-lymphoblastoid and yeast cells. Riluzole 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 9316860-1 1997 Cytochrome P450 (CYP) and uridine diphosphate glucuronosyltransferase (UGT) isoenzymes involved in riluzole oxidation and glucuronidation were characterized in (1) kinetic studies with human hepatic microsomes and isoenzyme-selective probes and (2) metabolic studies with genetically expressed human CYP isoenzymes from transfected B-lymphoblastoid and yeast cells. Riluzole 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 300-303 9322876-4 1997 The drugs both undergo hepatic metabolism; however, the specific cytochrome P-450 (CYP) enzymes responsible for hydroxylation are CYP 2C8 for paclitaxel and CYP 3A4 for docetaxel. Paclitaxel 142-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 9322876-4 1997 The drugs both undergo hepatic metabolism; however, the specific cytochrome P-450 (CYP) enzymes responsible for hydroxylation are CYP 2C8 for paclitaxel and CYP 3A4 for docetaxel. Paclitaxel 142-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 9322876-4 1997 The drugs both undergo hepatic metabolism; however, the specific cytochrome P-450 (CYP) enzymes responsible for hydroxylation are CYP 2C8 for paclitaxel and CYP 3A4 for docetaxel. Docetaxel 169-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 9322876-4 1997 The drugs both undergo hepatic metabolism; however, the specific cytochrome P-450 (CYP) enzymes responsible for hydroxylation are CYP 2C8 for paclitaxel and CYP 3A4 for docetaxel. Docetaxel 169-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 9350229-2 1997 The induction potential of metanil yellow on hepatic microsomal cytochrome P-450 (P-450)-dependent monooxygenases and cytosolic detoxification enzymes, namely, glutathione S-transferase (GST) and quinone reductase (QR), was investigated. metanil yellow 27-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 9242187-1 1997 Epoxyeicosatrienoic acids (EETs) are potent endothelium-derived vasodilators formed from cytochrome P-450 metabolism of arachidonic acid. epoxyeicosatrienoic acids 0-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 9242187-1 1997 Epoxyeicosatrienoic acids (EETs) are potent endothelium-derived vasodilators formed from cytochrome P-450 metabolism of arachidonic acid. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 9242187-1 1997 Epoxyeicosatrienoic acids (EETs) are potent endothelium-derived vasodilators formed from cytochrome P-450 metabolism of arachidonic acid. Arachidonic Acid 120-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 9247615-0 1997 Cytochrome P450 hydroxylation of carbon atoms of the alkyl chain of symmetrical N-nitrosodialkylamines by human liver microsomes. Carbon 33-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9247615-0 1997 Cytochrome P450 hydroxylation of carbon atoms of the alkyl chain of symmetrical N-nitrosodialkylamines by human liver microsomes. n-nitrosodialkylamines 80-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9223567-1 1997 The antihypertensive agent diltiazem (DTZ) impairs hepatic drug metabolism by inhibition of cytochrome P450 (CYP). Diltiazem 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 9248661-2 1997 Multiple forms of cytochrome P450 (CYP) catalyse the oxidation of chemicals of endogenous and exogenous origin, including drugs, carcinogens, steroids and eicosanoids. Steroids 142-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 9248661-2 1997 Multiple forms of cytochrome P450 (CYP) catalyse the oxidation of chemicals of endogenous and exogenous origin, including drugs, carcinogens, steroids and eicosanoids. Steroids 142-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 9248661-2 1997 Multiple forms of cytochrome P450 (CYP) catalyse the oxidation of chemicals of endogenous and exogenous origin, including drugs, carcinogens, steroids and eicosanoids. Eicosanoids 155-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 9248661-2 1997 Multiple forms of cytochrome P450 (CYP) catalyse the oxidation of chemicals of endogenous and exogenous origin, including drugs, carcinogens, steroids and eicosanoids. Eicosanoids 155-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 9248661-10 1997 Thus, drugs with side chains containing unsaturated carbon-carbon bonds and furan ring systems are associated with CYP inactivation. Carbon 52-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 9248661-10 1997 Thus, drugs with side chains containing unsaturated carbon-carbon bonds and furan ring systems are associated with CYP inactivation. Carbon 59-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 9248661-10 1997 Thus, drugs with side chains containing unsaturated carbon-carbon bonds and furan ring systems are associated with CYP inactivation. furan 76-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-118 9248661-11 1997 Nitrogen-containing systems may also inactivate CYP. Nitrogen 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 9248661-15 1997 Drugs that elicit CYP complexation include the first generation macrolide antibiotics, but newer analogues appear much safer. macrolide antibiotics 64-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-21 9223567-1 1997 The antihypertensive agent diltiazem (DTZ) impairs hepatic drug metabolism by inhibition of cytochrome P450 (CYP). Diltiazem 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-112 9223567-1 1997 The antihypertensive agent diltiazem (DTZ) impairs hepatic drug metabolism by inhibition of cytochrome P450 (CYP). Diltiazem 38-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 9223567-1 1997 The antihypertensive agent diltiazem (DTZ) impairs hepatic drug metabolism by inhibition of cytochrome P450 (CYP). Diltiazem 38-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-112 9223567-3 1997 Thus, DTZ metabolites may contribute to CYP inhibition. Diltiazem 6-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-43 9223567-4 1997 This study assessed the role of human CYPs in microsomal DTZ oxidation and the capacity of DTZ metabolites to inhibit specific CYP activities. Diltiazem 57-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 9241661-1 1997 Cyclophosphamide and ifosfamide are alkylating agent prodrugs that require activation by cytochrome P450 (CYP) to manifest their cancer chemotherapeutic activity. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 9253143-0 1997 Cytochrome P450 isozymes responsible for the metabolism of toluene and styrene in human liver microsomes. Toluene 59-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9253143-0 1997 Cytochrome P450 isozymes responsible for the metabolism of toluene and styrene in human liver microsomes. Styrene 71-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9253143-2 1997 Cytochrome P450 isozymes from Asian (31 Chinese subjects) and Caucasian (14 Finnish subjects) livers were examined for their roles in the metabolism of toluene (rates of benzyl alcohol, o- and p-cresol formation) and styrene (rates of styrene glycol formation). Toluene 152-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9253143-2 1997 Cytochrome P450 isozymes from Asian (31 Chinese subjects) and Caucasian (14 Finnish subjects) livers were examined for their roles in the metabolism of toluene (rates of benzyl alcohol, o- and p-cresol formation) and styrene (rates of styrene glycol formation). Benzyl Alcohol 170-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9253143-2 1997 Cytochrome P450 isozymes from Asian (31 Chinese subjects) and Caucasian (14 Finnish subjects) livers were examined for their roles in the metabolism of toluene (rates of benzyl alcohol, o- and p-cresol formation) and styrene (rates of styrene glycol formation). o- and p-cresol 186-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9253143-2 1997 Cytochrome P450 isozymes from Asian (31 Chinese subjects) and Caucasian (14 Finnish subjects) livers were examined for their roles in the metabolism of toluene (rates of benzyl alcohol, o- and p-cresol formation) and styrene (rates of styrene glycol formation). Styrene 217-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9253143-2 1997 Cytochrome P450 isozymes from Asian (31 Chinese subjects) and Caucasian (14 Finnish subjects) livers were examined for their roles in the metabolism of toluene (rates of benzyl alcohol, o- and p-cresol formation) and styrene (rates of styrene glycol formation). styrene glycol 235-249 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9255560-1 1997 Cytochrome P450 (CYP) enzymes catalyze the generation of reactive species capable of binding with cellular macromolecules, leading to acute and delayed toxicity. reactive species 57-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9255560-1 1997 Cytochrome P450 (CYP) enzymes catalyze the generation of reactive species capable of binding with cellular macromolecules, leading to acute and delayed toxicity. reactive species 57-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 9224780-0 1997 Evaluation of omeprazole and lansoprazole as inhibitors of cytochrome P450 isoforms. Omeprazole 14-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 9224780-0 1997 Evaluation of omeprazole and lansoprazole as inhibitors of cytochrome P450 isoforms. Lansoprazole 29-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 9224780-1 1997 The human clearance of omeprazole and lansoprazole is conducted primarily by the hepatic cytochrome P450 (CYP) system. Omeprazole 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 9224780-1 1997 The human clearance of omeprazole and lansoprazole is conducted primarily by the hepatic cytochrome P450 (CYP) system. Omeprazole 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 9224780-1 1997 The human clearance of omeprazole and lansoprazole is conducted primarily by the hepatic cytochrome P450 (CYP) system. Lansoprazole 38-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 9224780-1 1997 The human clearance of omeprazole and lansoprazole is conducted primarily by the hepatic cytochrome P450 (CYP) system. Lansoprazole 38-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 9193876-0 1997 Hydroxylation and demethylation of the tricyclic antidepressant nortriptyline by cDNA-expressed human cytochrome P-450 isozymes. Nortriptyline 64-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 9190854-0 1997 Reappraisal of human CYP isoforms involved in imipramine N-demethylation and 2-hydroxylation: a study using microsomes obtained from putative extensive and poor metabolizers of S-mephenytoin and eleven recombinant human CYPs. imipramine n 46-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 9190854-0 1997 Reappraisal of human CYP isoforms involved in imipramine N-demethylation and 2-hydroxylation: a study using microsomes obtained from putative extensive and poor metabolizers of S-mephenytoin and eleven recombinant human CYPs. Mephenytoin 177-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 9190854-1 1997 Cytochrome P450 (CYP) involved in the two major pathways of imipramine (IMI) was reappraised using human liver microsomes phenotyped for S-mephenytoin 4"-hydroxylation in vitro and 11 recombinant human CYP isoforms. Imipramine 60-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9190854-1 1997 Cytochrome P450 (CYP) involved in the two major pathways of imipramine (IMI) was reappraised using human liver microsomes phenotyped for S-mephenytoin 4"-hydroxylation in vitro and 11 recombinant human CYP isoforms. Imipramine 60-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 9190854-1 1997 Cytochrome P450 (CYP) involved in the two major pathways of imipramine (IMI) was reappraised using human liver microsomes phenotyped for S-mephenytoin 4"-hydroxylation in vitro and 11 recombinant human CYP isoforms. Imipramine 60-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-205 9190854-1 1997 Cytochrome P450 (CYP) involved in the two major pathways of imipramine (IMI) was reappraised using human liver microsomes phenotyped for S-mephenytoin 4"-hydroxylation in vitro and 11 recombinant human CYP isoforms. Imipramine 72-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9190854-1 1997 Cytochrome P450 (CYP) involved in the two major pathways of imipramine (IMI) was reappraised using human liver microsomes phenotyped for S-mephenytoin 4"-hydroxylation in vitro and 11 recombinant human CYP isoforms. Imipramine 72-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 9190854-1 1997 Cytochrome P450 (CYP) involved in the two major pathways of imipramine (IMI) was reappraised using human liver microsomes phenotyped for S-mephenytoin 4"-hydroxylation in vitro and 11 recombinant human CYP isoforms. Imipramine 72-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-205 9190854-1 1997 Cytochrome P450 (CYP) involved in the two major pathways of imipramine (IMI) was reappraised using human liver microsomes phenotyped for S-mephenytoin 4"-hydroxylation in vitro and 11 recombinant human CYP isoforms. Mephenytoin 137-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9190854-1 1997 Cytochrome P450 (CYP) involved in the two major pathways of imipramine (IMI) was reappraised using human liver microsomes phenotyped for S-mephenytoin 4"-hydroxylation in vitro and 11 recombinant human CYP isoforms. Mephenytoin 137-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 9187259-7 1997 Direct evidence for the in vivo epoxidation of arachidonic acid by intestinal cytochrome P450 was provided by documenting, for the first time, the presence of epoxyeicosatrienoic acids in human jejunum by gas chromatography/mass spectrometry. Arachidonic Acid 47-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 9187259-7 1997 Direct evidence for the in vivo epoxidation of arachidonic acid by intestinal cytochrome P450 was provided by documenting, for the first time, the presence of epoxyeicosatrienoic acids in human jejunum by gas chromatography/mass spectrometry. epoxyeicosatrienoic acids 159-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 9241661-1 1997 Cyclophosphamide and ifosfamide are alkylating agent prodrugs that require activation by cytochrome P450 (CYP) to manifest their cancer chemotherapeutic activity. Cyclophosphamide 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 9241661-1 1997 Cyclophosphamide and ifosfamide are alkylating agent prodrugs that require activation by cytochrome P450 (CYP) to manifest their cancer chemotherapeutic activity. Ifosfamide 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 9241661-1 1997 Cyclophosphamide and ifosfamide are alkylating agent prodrugs that require activation by cytochrome P450 (CYP) to manifest their cancer chemotherapeutic activity. Ifosfamide 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 9157990-8 1997 These studies: (a) demonstrate an underlying metabolic basis for the clinically important oxazaphosphorine autoinduction pharmacokinetics seen with these drugs in cancer patients; and (b) identify rifampin and other CYP inducers as potentially useful for increasing the rates of cyclophosphamide 4-hydroxylation and ifosfamide 4-hydroxylation in human liver in a manner that could favorably impact the clinical pharmacokinetics of these anticancer prodrugs. oxazaphosphorine 90-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-219 9212781-8 1997 Overall, our results indicate that the oxidative dehalo genation of para-halogenated phenols, resulting in the formation of benzoquinones, is not specific only for cytochrome P-450 enzymes. Phenols 85-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-180 9212781-8 1997 Overall, our results indicate that the oxidative dehalo genation of para-halogenated phenols, resulting in the formation of benzoquinones, is not specific only for cytochrome P-450 enzymes. Benzoquinones 124-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-180 9157990-1 1997 The anticancer oxazaphosphorine prodrugs cyclophosphamide and ifosfamide are activated in human liver by a 4-hydroxylation reaction catalyzed by multiple cytochrome P450 (CYP) enzymes. oxazaphosphorine 15-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 9157990-8 1997 These studies: (a) demonstrate an underlying metabolic basis for the clinically important oxazaphosphorine autoinduction pharmacokinetics seen with these drugs in cancer patients; and (b) identify rifampin and other CYP inducers as potentially useful for increasing the rates of cyclophosphamide 4-hydroxylation and ifosfamide 4-hydroxylation in human liver in a manner that could favorably impact the clinical pharmacokinetics of these anticancer prodrugs. Cyclophosphamide 279-295 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-219 9157990-1 1997 The anticancer oxazaphosphorine prodrugs cyclophosphamide and ifosfamide are activated in human liver by a 4-hydroxylation reaction catalyzed by multiple cytochrome P450 (CYP) enzymes. oxazaphosphorine 15-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-174 9157990-8 1997 These studies: (a) demonstrate an underlying metabolic basis for the clinically important oxazaphosphorine autoinduction pharmacokinetics seen with these drugs in cancer patients; and (b) identify rifampin and other CYP inducers as potentially useful for increasing the rates of cyclophosphamide 4-hydroxylation and ifosfamide 4-hydroxylation in human liver in a manner that could favorably impact the clinical pharmacokinetics of these anticancer prodrugs. Ifosfamide 316-326 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-219 9157990-1 1997 The anticancer oxazaphosphorine prodrugs cyclophosphamide and ifosfamide are activated in human liver by a 4-hydroxylation reaction catalyzed by multiple cytochrome P450 (CYP) enzymes. Cyclophosphamide 41-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 9157990-1 1997 The anticancer oxazaphosphorine prodrugs cyclophosphamide and ifosfamide are activated in human liver by a 4-hydroxylation reaction catalyzed by multiple cytochrome P450 (CYP) enzymes. Cyclophosphamide 41-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-174 9160172-6 1997 The most favoured explanation is that continuous tretinoin treatment acts to induce drug catabolism by cytochrome P450 (CYP) enzymes. Tretinoin 49-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 9157990-1 1997 The anticancer oxazaphosphorine prodrugs cyclophosphamide and ifosfamide are activated in human liver by a 4-hydroxylation reaction catalyzed by multiple cytochrome P450 (CYP) enzymes. Ifosfamide 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-169 9157990-1 1997 The anticancer oxazaphosphorine prodrugs cyclophosphamide and ifosfamide are activated in human liver by a 4-hydroxylation reaction catalyzed by multiple cytochrome P450 (CYP) enzymes. Ifosfamide 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 171-174 9176244-1 1997 We investigated the effect of 20-hydroxyeicosatetraenoic acid (20-HETE), an arachidonic acid metabolite of the cytochrome P-450 (cP450) 4A pathway, on human pulmonary arterial tone. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 30-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 9176244-1 1997 We investigated the effect of 20-hydroxyeicosatetraenoic acid (20-HETE), an arachidonic acid metabolite of the cytochrome P-450 (cP450) 4A pathway, on human pulmonary arterial tone. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 30-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-134 9176244-1 1997 We investigated the effect of 20-hydroxyeicosatetraenoic acid (20-HETE), an arachidonic acid metabolite of the cytochrome P-450 (cP450) 4A pathway, on human pulmonary arterial tone. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 9176244-1 1997 We investigated the effect of 20-hydroxyeicosatetraenoic acid (20-HETE), an arachidonic acid metabolite of the cytochrome P-450 (cP450) 4A pathway, on human pulmonary arterial tone. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-134 9160172-6 1997 The most favoured explanation is that continuous tretinoin treatment acts to induce drug catabolism by cytochrome P450 (CYP) enzymes. Tretinoin 49-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 9189656-12 1997 Argemone oil/alkaloid showed a Type II binding spectra with hepatic cytochrome P-450 (P-450) protein, thereby causing loss of P-450 content and an impairment of phase I and phase II enzymes. argemone oil 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 9130514-1 1997 The involvement of cytochrome-P450-dependent metabolism of small-molecular-weight compounds as a prerequisite for an optimal lymphocyte response to those compounds is discussed on the basis of studies with compound such as p-phenylenediamine, sulfamethoxazole, chloramphenicol and fragrances. 4-phenylenediamine 223-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 9189656-12 1997 Argemone oil/alkaloid showed a Type II binding spectra with hepatic cytochrome P-450 (P-450) protein, thereby causing loss of P-450 content and an impairment of phase I and phase II enzymes. Alkaloids 13-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 9130514-1 1997 The involvement of cytochrome-P450-dependent metabolism of small-molecular-weight compounds as a prerequisite for an optimal lymphocyte response to those compounds is discussed on the basis of studies with compound such as p-phenylenediamine, sulfamethoxazole, chloramphenicol and fragrances. Sulfamethoxazole 243-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 9130514-1 1997 The involvement of cytochrome-P450-dependent metabolism of small-molecular-weight compounds as a prerequisite for an optimal lymphocyte response to those compounds is discussed on the basis of studies with compound such as p-phenylenediamine, sulfamethoxazole, chloramphenicol and fragrances. Chloramphenicol 261-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 9152386-0 1997 Catalysis of the cysteine conjugation and protein binding of acetaminophen by microsomes from a human lymphoblast line transfected with the cDNAs of various forms of human cytochrome P450. Cysteine 17-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-187 9152386-0 1997 Catalysis of the cysteine conjugation and protein binding of acetaminophen by microsomes from a human lymphoblast line transfected with the cDNAs of various forms of human cytochrome P450. Acetaminophen 61-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-187 9152386-4 1997 This would indicate that some of forms of cytochrome P450 (CYP) catalyzing NABQI formation have their active site on the cytosolic surface and others on the lumenal surface. N-acetyl-4-benzoquinoneimine 75-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 9152386-4 1997 This would indicate that some of forms of cytochrome P450 (CYP) catalyzing NABQI formation have their active site on the cytosolic surface and others on the lumenal surface. N-acetyl-4-benzoquinoneimine 75-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 9226306-0 1997 Cytochrome P450 metabolites of arachidonic acid as intracellular signaling molecules in vascular tissue. Arachidonic Acid 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9041227-7 1997 Although the dogma is that the myoglobin is the source of iron, the results of recent studies suggest that cytochrome P-450 may be an important source of iron in this model. Iron 154-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-123 9070354-0 1997 Cytochrome P450-dependent metabolism of trichloroethylene: interindividual differences in humans. Trichloroethylene 40-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Ritonavir 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Ritonavir 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Saquinavir 113-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Saquinavir 113-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Indinavir 125-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Indinavir 125-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Nelfinavir 136-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. Nelfinavir 136-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. amprenavir 152-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 9056009-2 1997 In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. amprenavir 152-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 9056009-3 1997 The structural features of ritonavir responsible for CYP binding and inhibition were examined. Ritonavir 27-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-56 27414767-6 1997 Cytochrome P-450 enzymes can hydroxylate salicylate to produce 2,5-DHBA, and it is likely that phenylalanine hydroxylase produces most of the p-tyrosine detected in hepatic tissues. Salicylates 41-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 27414767-6 1997 Cytochrome P-450 enzymes can hydroxylate salicylate to produce 2,5-DHBA, and it is likely that phenylalanine hydroxylase produces most of the p-tyrosine detected in hepatic tissues. 2,5-dihydroxybenzoic acid 63-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 27414767-6 1997 Cytochrome P-450 enzymes can hydroxylate salicylate to produce 2,5-DHBA, and it is likely that phenylalanine hydroxylase produces most of the p-tyrosine detected in hepatic tissues. Tyrosine 142-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 9130383-2 1997 It was discovered that a long wave shift occurred in the fluorescence spectra of cytochrome P450, both in solution and within phospholipid vesicles. Phospholipids 126-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 9130383-3 1997 The efficiency of quenching of membrane-bound cytochrome P450 with acrylamide also increased after freezing. Acrylamide 67-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 9078706-0 1997 [Alcohol oxidative metabolism systems of microsomes (cytochrome P-450)]. Alcohols 1-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-71 9070354-2 1997 Cytochrome P450 (CYP)-dependent metabolism of TRI produces chloral hydrate (CH) and is rate limiting in the ultimate production of trichloro- and/or dichloroacetic acid from TRI. Chloral Hydrate 59-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9070354-2 1997 Cytochrome P450 (CYP)-dependent metabolism of TRI produces chloral hydrate (CH) and is rate limiting in the ultimate production of trichloro- and/or dichloroacetic acid from TRI. Chloral Hydrate 59-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 9070354-2 1997 Cytochrome P450 (CYP)-dependent metabolism of TRI produces chloral hydrate (CH) and is rate limiting in the ultimate production of trichloro- and/or dichloroacetic acid from TRI. trichloro- 131-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9070354-2 1997 Cytochrome P450 (CYP)-dependent metabolism of TRI produces chloral hydrate (CH) and is rate limiting in the ultimate production of trichloro- and/or dichloroacetic acid from TRI. trichloro- 131-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 9070354-2 1997 Cytochrome P450 (CYP)-dependent metabolism of TRI produces chloral hydrate (CH) and is rate limiting in the ultimate production of trichloro- and/or dichloroacetic acid from TRI. Dichloroacetic Acid 149-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 9070354-2 1997 Cytochrome P450 (CYP)-dependent metabolism of TRI produces chloral hydrate (CH) and is rate limiting in the ultimate production of trichloro- and/or dichloroacetic acid from TRI. Dichloroacetic Acid 149-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 9003190-1 1997 Cytochrome P450 (P450) 3A4 is the most abundant human P450 and oxidizes a diversity of substrates, including various drugs, steroids, carcinogens, and macrolide natural products. Steroids 124-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 15374139-3 1997 Although this system is an important component of the defenses that protect living organisms against toxic chemicals, some reactions catalyzed by the cytochrome P-450 system result in the formation of products that are highly reactive as well as active oxygen species. Oxygen 253-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 8995391-4 1997 Classical cytochrome P-450 ligands such as the mechanism-based inactivator 1-aminobenzotriazole did not inhibit iNOS. 1-aminobenzotriazole 75-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 8995391-7 1997 The binding of chlorzoxazone to iNOS and human and rat liver microsomal cytochrome P-450 induced a high spin, type I spectra, which was reversed by imidazole. Chlorzoxazone 15-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 8995391-7 1997 The binding of chlorzoxazone to iNOS and human and rat liver microsomal cytochrome P-450 induced a high spin, type I spectra, which was reversed by imidazole. imidazole 148-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 9010633-0 1997 Identification of rat and human cytochrome P450 forms involved in the metabolism of the thromboxane A2 receptor antagonist (+)-S-145. S 145 123-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 9443608-13 1997 CONCLUSIONS: Halomon is metabolized by mouse and human hepatic cytochrome P-450 enzymes, the identities of which remain unknown. 6-bromo-3-(bromomethyl)-7-methyl-2,3,7-trichloro-1-octene 13-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 9054609-1 1997 Several naturally occurring coumarins to which humans are routinely exposed have been previously found to be potent inhibitors and inactivators of cytochrome P450 (P450) 1A1-mediated monooxygenase in both murine hepatic microsomes and in a reconstituted system using purified human P450 1A1 [Cai et al. Coumarins 28-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-173 9068933-5 1997 This suggests that the change in tolbutamide clearance may be due to a slight inhibition of the cytochrome P450 (CYP) isoenzyme CYP2C9/10 when sertraline was administered in its maximum recommended dosage. Tolbutamide 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-111 9068933-5 1997 This suggests that the change in tolbutamide clearance may be due to a slight inhibition of the cytochrome P450 (CYP) isoenzyme CYP2C9/10 when sertraline was administered in its maximum recommended dosage. Tolbutamide 33-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 9068933-5 1997 This suggests that the change in tolbutamide clearance may be due to a slight inhibition of the cytochrome P450 (CYP) isoenzyme CYP2C9/10 when sertraline was administered in its maximum recommended dosage. Sertraline 143-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-111 9068933-5 1997 This suggests that the change in tolbutamide clearance may be due to a slight inhibition of the cytochrome P450 (CYP) isoenzyme CYP2C9/10 when sertraline was administered in its maximum recommended dosage. Sertraline 143-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-116 9068934-8 1997 The metabolism of warfarin is principally mediated by the cytochrome P450 (CYP) isoenzyme CYP2C9/10. Warfarin 18-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 9068934-8 1997 The metabolism of warfarin is principally mediated by the cytochrome P450 (CYP) isoenzyme CYP2C9/10. Warfarin 18-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-78 9010633-2 1997 We investigated the oxidative metabolism of (+)-S-145 and its beta-oxidized metabolites with liver microsomes from rats and humans to identify which cytochrome P450 (P450) forms are involved in these reactions. S 145 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-164 9048233-5 1997 Metanil yellow treatment for 7 days also led to a significant increase in cytochrome P-450 (P-450)-dependent aryl hydrocarbon hydroxylase (AHH) activity (99-223%) in the liver and intestine. metanil yellow 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 9342584-0 1997 Metabolism of carteolol by cDNA-expressed human cytochrome P450. Carteolol 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 9342584-1 1997 OBJECTIVES: To determine human cytochrome P450 isoform(s) (CYPs) involved in the metabolism of carteolol, the biotransformation of the compound was investigated in vitro using ten isoforms of human cytochrome P450 expressed in human AHH-1 TK +/- cell lines. Carteolol 95-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 9342584-1 1997 OBJECTIVES: To determine human cytochrome P450 isoform(s) (CYPs) involved in the metabolism of carteolol, the biotransformation of the compound was investigated in vitro using ten isoforms of human cytochrome P450 expressed in human AHH-1 TK +/- cell lines. Carteolol 95-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-213 9342584-8 1997 CONCLUSION: 8-Hydroxylation is the only cytochrome P450-catalyzed metabolic reaction of carteolol by its expressed microsomes, and CYP2D6 is the principal isoform of the enzyme involved in the catalytic reaction. Carteolol 88-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 9279132-4 1997 Liver microsomal cytochrome P-450 level was found to be low in ascorbate deficient animals. Ascorbic Acid 63-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 9046019-0 1997 NADPH-initiated cytochrome P450-dependent free iron-independent microsomal lipid peroxidation: specific prevention by ascorbic acid. NADP 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 9046019-0 1997 NADPH-initiated cytochrome P450-dependent free iron-independent microsomal lipid peroxidation: specific prevention by ascorbic acid. Iron 47-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 9046019-0 1997 NADPH-initiated cytochrome P450-dependent free iron-independent microsomal lipid peroxidation: specific prevention by ascorbic acid. Ascorbic Acid 118-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 9046019-1 1997 In this paper we demonstrate that ascorbic acid specifically prevents NADPH-initiated cytochrome P450 (P450)-mediated microsomal lipid peroxidation in the absence of free iron. Ascorbic Acid 34-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 9046019-1 1997 In this paper we demonstrate that ascorbic acid specifically prevents NADPH-initiated cytochrome P450 (P450)-mediated microsomal lipid peroxidation in the absence of free iron. NADP 70-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-101 9364799-1 1997 Arachidonic acid (AA) can be metabolized to a variety of lipid mediators including prostaglandins (PGE), and hydroxyeicosatetraenoic acids (HETE) by cyclooxygenase, lipoxygenase and cytochrome P450-dependent monooxygenase enzymatic pathways. Hydroxyeicosatetraenoic Acids 109-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-197 9200152-7 1997 In summary, our findings suggested that dietary menhaden oil can modulate ADH and CYP activities in a manner that may alter the metabolism of CP and, therefore, improve its therapeutic index by increasing its therapeutic effect and decreasing its toxicity. Menhaden oil 48-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-85 9364799-1 1997 Arachidonic acid (AA) can be metabolized to a variety of lipid mediators including prostaglandins (PGE), and hydroxyeicosatetraenoic acids (HETE) by cyclooxygenase, lipoxygenase and cytochrome P450-dependent monooxygenase enzymatic pathways. Hydroxyeicosatetraenoic Acids 140-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-197 9364799-1 1997 Arachidonic acid (AA) can be metabolized to a variety of lipid mediators including prostaglandins (PGE), and hydroxyeicosatetraenoic acids (HETE) by cyclooxygenase, lipoxygenase and cytochrome P450-dependent monooxygenase enzymatic pathways. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-197 8971135-2 1996 The aim of this study was to identify the cytochrome P450 (CYP) enzyme primarily responsible for the NADPH-dependent covalent binding of [14C]halothane to human liver microsomes. NADP 101-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 8975714-0 1996 Structure and mapping of the human lanosterol 14alpha-demethylase gene (CYP51) encoding the cytochrome P450 involved in cholesterol biosynthesis; comparison of exon/intron organization with other mammalian and fungal CYP genes. Cholesterol 120-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-107 8975714-0 1996 Structure and mapping of the human lanosterol 14alpha-demethylase gene (CYP51) encoding the cytochrome P450 involved in cholesterol biosynthesis; comparison of exon/intron organization with other mammalian and fungal CYP genes. Cholesterol 120-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-75 8975714-1 1996 Sterol 14alpha-demethylase (P45014DM) encoded by CYP51 is a member of the cytochrome P450 (CYP) gene superfamily involved in sterol biosynthesis in fungi, plants, and animals. Sterols 125-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 8975714-1 1996 Sterol 14alpha-demethylase (P45014DM) encoded by CYP51 is a member of the cytochrome P450 (CYP) gene superfamily involved in sterol biosynthesis in fungi, plants, and animals. Sterols 125-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 8971135-2 1996 The aim of this study was to identify the cytochrome P450 (CYP) enzyme primarily responsible for the NADPH-dependent covalent binding of [14C]halothane to human liver microsomes. [14c]halothane 137-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 8971135-2 1996 The aim of this study was to identify the cytochrome P450 (CYP) enzyme primarily responsible for the NADPH-dependent covalent binding of [14C]halothane to human liver microsomes. NADP 101-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 8971135-2 1996 The aim of this study was to identify the cytochrome P450 (CYP) enzyme primarily responsible for the NADPH-dependent covalent binding of [14C]halothane to human liver microsomes. [14c]halothane 137-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 9030997-4 1996 Cytochrome P-450 and other enzymes involved in aldosterone synthesis were found in the tumor portions but not in the zona glomerulosa of attached adrenals, which histopathologically showed "paradoxical hyperplasia". Aldosterone 47-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8942676-0 1996 Role of the polarity of the heme environment for the CO stretch modes in cytochrome P-450cam-CO. Heme 28-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-95 8944409-8 1996 The increased buspirone concentrations are likely to be due to inhibition of first pass liver metabolism by fluvoxamine acting on the cytochrome P-450 system. Buspirone 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 8916898-5 1996 In the reduced, CO-bound form of cytochrome P-450cam at 290 K the (1S)-camphor complex reveals another CO stretch vibration population distribution with slightly higher frequencies [1940.2 cm-1 (major band) and 1946.3 cm-1 (minor band)] compared to the (1R)-camphor complex [1939.7 cm-1 (major band) and 1930 cm-1 (minor band)]. Camphor 258-265 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 8916898-7 1996 Assuming the carbon monoxide complex as a model for the dioxygen complex, the more loosened binding of (1S)-camphor, therefore the increased water accessibility, and the weaker contact of the iron ligand to the I-helix might explain the higher amount of uncoupling of the cytochrome P-450 reaction cycle compared to that when (1R)-camphor is used as substrate. Carbon Monoxide 13-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 272-288 8916898-7 1996 Assuming the carbon monoxide complex as a model for the dioxygen complex, the more loosened binding of (1S)-camphor, therefore the increased water accessibility, and the weaker contact of the iron ligand to the I-helix might explain the higher amount of uncoupling of the cytochrome P-450 reaction cycle compared to that when (1R)-camphor is used as substrate. Oxygen 56-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 272-288 8937855-2 1996 In this study, human cytochrome P450 (CYP) isoform involved in cotinine formation was identified. Cotinine 63-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 8937855-2 1996 In this study, human cytochrome P450 (CYP) isoform involved in cotinine formation was identified. Cotinine 63-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 8940291-1 1996 As a participant of the endogenous arachidonic acid metabolic cascade, microsomal cytochrome P450 metabolizes the fatty acid to biologically active hydroxyeicosatetraenoic and epoxyeicosatrienoic acids. Arachidonic Acid 35-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 8940291-1 1996 As a participant of the endogenous arachidonic acid metabolic cascade, microsomal cytochrome P450 metabolizes the fatty acid to biologically active hydroxyeicosatetraenoic and epoxyeicosatrienoic acids. Fatty Acids 114-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 8940291-1 1996 As a participant of the endogenous arachidonic acid metabolic cascade, microsomal cytochrome P450 metabolizes the fatty acid to biologically active hydroxyeicosatetraenoic and epoxyeicosatrienoic acids. hydroxyeicosatetraenoic 148-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 8940291-1 1996 As a participant of the endogenous arachidonic acid metabolic cascade, microsomal cytochrome P450 metabolizes the fatty acid to biologically active hydroxyeicosatetraenoic and epoxyeicosatrienoic acids. epoxyeicosatrienoic acids 176-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 8916898-0 1996 Structural changes in cytochrome P-450cam effected by the binding of the enantiomers (1R)-camphor and (1S)-camphor. Camphor 85-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 8916898-0 1996 Structural changes in cytochrome P-450cam effected by the binding of the enantiomers (1R)-camphor and (1S)-camphor. Camphor 102-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 8916898-4 1996 The half-transition temperature of the thermal unfolding of 53.8 degrees C for the (1S)-camphor-bound oxidized cytochrome P-450cam is one degree lower than the value for the (1R)-camphor-bound protein (54.8 degrees C). Camphor 88-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 8916898-4 1996 The half-transition temperature of the thermal unfolding of 53.8 degrees C for the (1S)-camphor-bound oxidized cytochrome P-450cam is one degree lower than the value for the (1R)-camphor-bound protein (54.8 degrees C). Camphor 179-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 8916898-5 1996 In the reduced, CO-bound form of cytochrome P-450cam at 290 K the (1S)-camphor complex reveals another CO stretch vibration population distribution with slightly higher frequencies [1940.2 cm-1 (major band) and 1946.3 cm-1 (minor band)] compared to the (1R)-camphor complex [1939.7 cm-1 (major band) and 1930 cm-1 (minor band)]. (1s) 66-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 8916898-5 1996 In the reduced, CO-bound form of cytochrome P-450cam at 290 K the (1S)-camphor complex reveals another CO stretch vibration population distribution with slightly higher frequencies [1940.2 cm-1 (major band) and 1946.3 cm-1 (minor band)] compared to the (1R)-camphor complex [1939.7 cm-1 (major band) and 1930 cm-1 (minor band)]. Camphor 71-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 8951176-0 1996 Oxidative metabolism of flunarizine and cinnarizine by microsomes from B-lymphoblastoid cell lines expressing human cytochrome P450 enzymes. Flunarizine 24-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 8951176-0 1996 Oxidative metabolism of flunarizine and cinnarizine by microsomes from B-lymphoblastoid cell lines expressing human cytochrome P450 enzymes. Cinnarizine 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-131 8970575-5 1996 EDHF may be a cytochrome P-450 metabolite of arachidonic acid. edhf 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 8970575-5 1996 EDHF may be a cytochrome P-450 metabolite of arachidonic acid. Arachidonic Acid 45-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 8944409-8 1996 The increased buspirone concentrations are likely to be due to inhibition of first pass liver metabolism by fluvoxamine acting on the cytochrome P-450 system. Fluvoxamine 108-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-150 8952228-6 1996 A process known to generate the O2 is the induction of certain cytochrome P450 (CYP) isozymes by drugs or environmental pollutants. Superoxides 32-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 8948090-0 1996 Effects of roxithromycin, erythromycin and troleandomycin on their N-demethylation by rat and human cytochrome P450 enzymes. Roxithromycin 11-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 8948090-0 1996 Effects of roxithromycin, erythromycin and troleandomycin on their N-demethylation by rat and human cytochrome P450 enzymes. Erythromycin 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 8948090-0 1996 Effects of roxithromycin, erythromycin and troleandomycin on their N-demethylation by rat and human cytochrome P450 enzymes. Troleandomycin 43-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-115 8952228-6 1996 A process known to generate the O2 is the induction of certain cytochrome P450 (CYP) isozymes by drugs or environmental pollutants. Superoxides 32-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-83 8952228-7 1996 RESULTS: We report: 1) a correlation between the induction of each CYP gene family and the O2 yield; 2) support to an observation reported previously that the tumor promoting ability of CYP inducers is mainly mediated by the O2; and 3) the description of a method for nitroxide mediated O2 detection in vivo. Superoxides 91-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 8952228-7 1996 RESULTS: We report: 1) a correlation between the induction of each CYP gene family and the O2 yield; 2) support to an observation reported previously that the tumor promoting ability of CYP inducers is mainly mediated by the O2; and 3) the description of a method for nitroxide mediated O2 detection in vivo. Superoxides 91-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-189 8952228-7 1996 RESULTS: We report: 1) a correlation between the induction of each CYP gene family and the O2 yield; 2) support to an observation reported previously that the tumor promoting ability of CYP inducers is mainly mediated by the O2; and 3) the description of a method for nitroxide mediated O2 detection in vivo. Superoxides 225-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 8952228-7 1996 RESULTS: We report: 1) a correlation between the induction of each CYP gene family and the O2 yield; 2) support to an observation reported previously that the tumor promoting ability of CYP inducers is mainly mediated by the O2; and 3) the description of a method for nitroxide mediated O2 detection in vivo. Superoxides 225-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-189 8952228-7 1996 RESULTS: We report: 1) a correlation between the induction of each CYP gene family and the O2 yield; 2) support to an observation reported previously that the tumor promoting ability of CYP inducers is mainly mediated by the O2; and 3) the description of a method for nitroxide mediated O2 detection in vivo. Hydroxylamine 268-277 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 8952228-7 1996 RESULTS: We report: 1) a correlation between the induction of each CYP gene family and the O2 yield; 2) support to an observation reported previously that the tumor promoting ability of CYP inducers is mainly mediated by the O2; and 3) the description of a method for nitroxide mediated O2 detection in vivo. Hydroxylamine 268-277 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-189 8952228-7 1996 RESULTS: We report: 1) a correlation between the induction of each CYP gene family and the O2 yield; 2) support to an observation reported previously that the tumor promoting ability of CYP inducers is mainly mediated by the O2; and 3) the description of a method for nitroxide mediated O2 detection in vivo. Superoxides 225-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-70 8952228-7 1996 RESULTS: We report: 1) a correlation between the induction of each CYP gene family and the O2 yield; 2) support to an observation reported previously that the tumor promoting ability of CYP inducers is mainly mediated by the O2; and 3) the description of a method for nitroxide mediated O2 detection in vivo. Superoxides 225-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 186-189 8964509-1 1996 Thromboxane synthase (TS) is a cytochrome P-450 (CYP450) enzyme catalyzing the conversion of prostaglandin endoperoxide (PGH2) into thromboxane A2 (TxA2) which plays a crucial role in hemostasis and cardiovascular diseases. Prostaglandin Endoperoxides 93-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 8946477-0 1996 CYP2D6 is the principal cytochrome P450 responsible for metabolism of the histamine H1 antagonist promethazine in human liver microsomes. Histamine 74-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 8946477-0 1996 CYP2D6 is the principal cytochrome P450 responsible for metabolism of the histamine H1 antagonist promethazine in human liver microsomes. Promethazine 98-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 8946477-1 1996 To determine which cytochrome P450 form is involved in the promethazine [10-(2-dimethylaminopropyl) phenothiazine] metabolism, in vitro analysis using human liver microsomes were performed. Promethazine 59-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 8946477-1 1996 To determine which cytochrome P450 form is involved in the promethazine [10-(2-dimethylaminopropyl) phenothiazine] metabolism, in vitro analysis using human liver microsomes were performed. Promethazine 73-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 8823154-0 1996 Involvement of alcohol dehydrogenase, short-chain dehydrogenase/reductase, aldehyde dehydrogenase, and cytochrome P450 in the control of retinoid signaling by activation of retinoic acid synthesis. Retinoids 137-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 8823154-0 1996 Involvement of alcohol dehydrogenase, short-chain dehydrogenase/reductase, aldehyde dehydrogenase, and cytochrome P450 in the control of retinoid signaling by activation of retinoic acid synthesis. Tretinoin 173-186 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 8823154-5 1996 Members of the alcohol dehydrogenase and short-chain dehydrogenase/reductase enzyme families catalyze the reversible interconversion of retinol and retinal, the rate-limiting step, whereas members of the aldehyde dehydrogenase and cytochrome P450 enzyme families catalyze the irreversible oxidation of retinal to retinoic acid. Vitamin A 136-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 231-246 8823154-5 1996 Members of the alcohol dehydrogenase and short-chain dehydrogenase/reductase enzyme families catalyze the reversible interconversion of retinol and retinal, the rate-limiting step, whereas members of the aldehyde dehydrogenase and cytochrome P450 enzyme families catalyze the irreversible oxidation of retinal to retinoic acid. Aldehydes 204-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 231-246 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 111-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 8964509-1 1996 Thromboxane synthase (TS) is a cytochrome P-450 (CYP450) enzyme catalyzing the conversion of prostaglandin endoperoxide (PGH2) into thromboxane A2 (TxA2) which plays a crucial role in hemostasis and cardiovascular diseases. Prostaglandin Endoperoxides 93-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-55 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 111-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 111-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-237 8964509-1 1996 Thromboxane synthase (TS) is a cytochrome P-450 (CYP450) enzyme catalyzing the conversion of prostaglandin endoperoxide (PGH2) into thromboxane A2 (TxA2) which plays a crucial role in hemostasis and cardiovascular diseases. Prostaglandin H2 121-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 111-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-237 8964509-1 1996 Thromboxane synthase (TS) is a cytochrome P-450 (CYP450) enzyme catalyzing the conversion of prostaglandin endoperoxide (PGH2) into thromboxane A2 (TxA2) which plays a crucial role in hemostasis and cardiovascular diseases. Prostaglandin H2 121-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-55 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 125-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 125-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 125-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-237 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 125-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 234-237 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 286-298 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 8765473-1 1996 Various complementary approaches were used to elucidate the major cytochrome P450 (CYP) enzyme responsible for mifepristone (RU 486) demethylation and hydroxylation in human liver microsomes: chemical and immunoinhibition of specific CYPs; correlation analyses between initial rates of mifepristone metabolism and relative immunodetectable CYP levels and rates of CYP marker substrate metabolism; and evaluation of metabolism by cDNA-expressed CYP3A4. Mifepristone 286-298 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-86 8964509-1 1996 Thromboxane synthase (TS) is a cytochrome P-450 (CYP450) enzyme catalyzing the conversion of prostaglandin endoperoxide (PGH2) into thromboxane A2 (TxA2) which plays a crucial role in hemostasis and cardiovascular diseases. Thromboxane A2 132-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 8964509-1 1996 Thromboxane synthase (TS) is a cytochrome P-450 (CYP450) enzyme catalyzing the conversion of prostaglandin endoperoxide (PGH2) into thromboxane A2 (TxA2) which plays a crucial role in hemostasis and cardiovascular diseases. Thromboxane A2 132-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-55 8884182-1 1996 Liarozole inhibits cytochrome P-450-dependent enzymes that play a key role in all-trans-retinoic acid (ATRA) catabolism. liarozole 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 8824525-0 1996 Cytochrome P450 species involved in the metabolism of quinoline. quinoline 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8824525-2 1996 The specific cytochrome P450 enzymes involved in quinoline metabolism in human and rat liver microsomes were determined using cDNA-expressed cytochrome P450s, correlations with specific cytochrome P450-linked monooxygenase activities in human liver microsomes and inhibition by specific inhibitors and antibodies. quinoline 49-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 8824525-2 1996 The specific cytochrome P450 enzymes involved in quinoline metabolism in human and rat liver microsomes were determined using cDNA-expressed cytochrome P450s, correlations with specific cytochrome P450-linked monooxygenase activities in human liver microsomes and inhibition by specific inhibitors and antibodies. quinoline 49-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-156 8824525-3 1996 CYP2A6 is the principal cytochrome P450 involved in the formation of quinoline-1-oxide in human liver microsomes (correlation coefficient r = 0.95), but is formed in only minute quantities in rat liver microsomes. Quinoline 1-oxide 69-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 8824525-4 1996 CYP2E1 is the principal cytochrome P450 involved in the formation of 3-hydroxyquinoline (r = 0.93) in human liver microsomes and is involved in the formation in rat liver microsomes. 3-Hydroxyquinoline 69-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 8824531-0 1996 Cytochrome P450 metabolic dealkylation of nine N-nitrosodialkylamines by human liver microsomes. n-nitrosodialkylamines 47-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8824531-3 1996 As the length of the alkyl chain increased from methyl to pentyl, dealkylation of symmetrical nitrosodialkylamines became less efficiently catalyzed by cytochrome P450. nitrosodialkylamines 94-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-167 8886602-1 1996 Midazolam (MDZ) is metabolized in human liver microsomes by the cytochrome P450 (CYP) 3A subfamily to 1"-hydroxy (1"-OH) and 4-hydroxy (4-OH) metabolites. Midazolam 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 8886602-1 1996 Midazolam (MDZ) is metabolized in human liver microsomes by the cytochrome P450 (CYP) 3A subfamily to 1"-hydroxy (1"-OH) and 4-hydroxy (4-OH) metabolites. Midazolam 11-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 8886602-1 1996 Midazolam (MDZ) is metabolized in human liver microsomes by the cytochrome P450 (CYP) 3A subfamily to 1"-hydroxy (1"-OH) and 4-hydroxy (4-OH) metabolites. 1"-hydroxy 102-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 8886602-1 1996 Midazolam (MDZ) is metabolized in human liver microsomes by the cytochrome P450 (CYP) 3A subfamily to 1"-hydroxy (1"-OH) and 4-hydroxy (4-OH) metabolites. 1"-oh 114-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 8886602-1 1996 Midazolam (MDZ) is metabolized in human liver microsomes by the cytochrome P450 (CYP) 3A subfamily to 1"-hydroxy (1"-OH) and 4-hydroxy (4-OH) metabolites. hydroxide ion 105-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 8886602-1 1996 Midazolam (MDZ) is metabolized in human liver microsomes by the cytochrome P450 (CYP) 3A subfamily to 1"-hydroxy (1"-OH) and 4-hydroxy (4-OH) metabolites. 4-oh 136-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-79 8877251-10 1996 In other tissues low in CYP activity, prostaglandin H synthase may also be responsible for bioactivation; e.g. in the kidney paracetamol (acetaminophen) toxicity is though to result from activation via this enzyme. Acetaminophen 125-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 8877251-10 1996 In other tissues low in CYP activity, prostaglandin H synthase may also be responsible for bioactivation; e.g. in the kidney paracetamol (acetaminophen) toxicity is though to result from activation via this enzyme. Acetaminophen 138-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 8884182-1 1996 Liarozole inhibits cytochrome P-450-dependent enzymes that play a key role in all-trans-retinoic acid (ATRA) catabolism. Tretinoin 82-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 8884182-1 1996 Liarozole inhibits cytochrome P-450-dependent enzymes that play a key role in all-trans-retinoic acid (ATRA) catabolism. Tretinoin 103-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 21541530-1 1996 To examine the possibility that chemical inducers of cytochrome P450 may have effects on the expression of oncogenes and a tumor suppressor gene, human colon LS174T cells were treated with different cytochrome P450 inducers such as benzanthracene (Ba), pyrazole (Pyr) and phenobarbital-hydrocortisone (Pb-Hc). benz(a)anthracene 232-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 21541530-1 1996 To examine the possibility that chemical inducers of cytochrome P450 may have effects on the expression of oncogenes and a tumor suppressor gene, human colon LS174T cells were treated with different cytochrome P450 inducers such as benzanthracene (Ba), pyrazole (Pyr) and phenobarbital-hydrocortisone (Pb-Hc). benz(a)anthracene 248-250 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 21541530-1 1996 To examine the possibility that chemical inducers of cytochrome P450 may have effects on the expression of oncogenes and a tumor suppressor gene, human colon LS174T cells were treated with different cytochrome P450 inducers such as benzanthracene (Ba), pyrazole (Pyr) and phenobarbital-hydrocortisone (Pb-Hc). pyrazole 253-261 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 21541530-1 1996 To examine the possibility that chemical inducers of cytochrome P450 may have effects on the expression of oncogenes and a tumor suppressor gene, human colon LS174T cells were treated with different cytochrome P450 inducers such as benzanthracene (Ba), pyrazole (Pyr) and phenobarbital-hydrocortisone (Pb-Hc). pyrazole 263-266 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 21541530-1 1996 To examine the possibility that chemical inducers of cytochrome P450 may have effects on the expression of oncogenes and a tumor suppressor gene, human colon LS174T cells were treated with different cytochrome P450 inducers such as benzanthracene (Ba), pyrazole (Pyr) and phenobarbital-hydrocortisone (Pb-Hc). phenobarbital-hydrocortisone 272-300 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 21541530-1 1996 To examine the possibility that chemical inducers of cytochrome P450 may have effects on the expression of oncogenes and a tumor suppressor gene, human colon LS174T cells were treated with different cytochrome P450 inducers such as benzanthracene (Ba), pyrazole (Pyr) and phenobarbital-hydrocortisone (Pb-Hc). pb-hc 302-307 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 8864312-11 1996 These in vitro data provide direct evidence for cytochrome P450 inhibition as the mechanism for the well documented diazepam-omeprazole clinical interaction and indicate that omeprazole sulphone, as well as the parent drug, contribute to the inhibition effect. diazepam-omeprazole 116-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 8765221-0 1996 Interaction of nitrogen monoxide with cytochrome P-450 monitored by surface-enhanced resonance Raman scattering. Nitric Oxide 15-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 8765221-1 1996 The reaction of mammalian cytochrome P-450 2B4 with nitrogen monoxide and oxygen has been studied by surface-enhanced resonance Raman scattering (SERRS) to obtain sharp and definitive information in situ on the nature of the changes in the active site pocket. Nitric Oxide 52-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 8765221-1 1996 The reaction of mammalian cytochrome P-450 2B4 with nitrogen monoxide and oxygen has been studied by surface-enhanced resonance Raman scattering (SERRS) to obtain sharp and definitive information in situ on the nature of the changes in the active site pocket. Oxygen 74-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 8827397-4 1996 Omeprazole, lansoprazole and pantoprazole are all mainly metabolished by the polymorphically expressed cytochrome P450 (CYP) isoform S-mephenytoin hydroxylase (CYP2C19), which means that within a population a few individuals (3% of Caucasians) metabolise the compounds slowly compared with the majority of the population. Omeprazole 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 8694760-2 1996 produced during the catalytic activity of nitric oxide synthase (NOS) and cytochrome P-450 has been implicated in the oxidative denitrification of hydroxyguanidines ( > C = NOH). hydroxyguanidine 147-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 8712747-10 1996 Itraconazole and its analogues are inhibitors of both cytochrome P450 and lipoxygenase and since itraconazole can modulate BU pharmacokinetics, oxidative catabolism is probably a determinant of BU metabolism. Itraconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-86 8827397-4 1996 Omeprazole, lansoprazole and pantoprazole are all mainly metabolished by the polymorphically expressed cytochrome P450 (CYP) isoform S-mephenytoin hydroxylase (CYP2C19), which means that within a population a few individuals (3% of Caucasians) metabolise the compounds slowly compared with the majority of the population. Omeprazole 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 8827397-4 1996 Omeprazole, lansoprazole and pantoprazole are all mainly metabolished by the polymorphically expressed cytochrome P450 (CYP) isoform S-mephenytoin hydroxylase (CYP2C19), which means that within a population a few individuals (3% of Caucasians) metabolise the compounds slowly compared with the majority of the population. Lansoprazole 12-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 8827397-4 1996 Omeprazole, lansoprazole and pantoprazole are all mainly metabolished by the polymorphically expressed cytochrome P450 (CYP) isoform S-mephenytoin hydroxylase (CYP2C19), which means that within a population a few individuals (3% of Caucasians) metabolise the compounds slowly compared with the majority of the population. Lansoprazole 12-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 8827397-4 1996 Omeprazole, lansoprazole and pantoprazole are all mainly metabolished by the polymorphically expressed cytochrome P450 (CYP) isoform S-mephenytoin hydroxylase (CYP2C19), which means that within a population a few individuals (3% of Caucasians) metabolise the compounds slowly compared with the majority of the population. Pantoprazole 29-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 8827397-4 1996 Omeprazole, lansoprazole and pantoprazole are all mainly metabolished by the polymorphically expressed cytochrome P450 (CYP) isoform S-mephenytoin hydroxylase (CYP2C19), which means that within a population a few individuals (3% of Caucasians) metabolise the compounds slowly compared with the majority of the population. Pantoprazole 29-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-123 8827397-12 1996 The effect of the acid pump inhibitors on enzymes in the liver has been intensely debated, and some authors have claimed that lansoprazole and pantoprazole have less potential than omeprazole to interact with other drugs metabolised by CYP. Lansoprazole 126-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 236-239 8827397-12 1996 The effect of the acid pump inhibitors on enzymes in the liver has been intensely debated, and some authors have claimed that lansoprazole and pantoprazole have less potential than omeprazole to interact with other drugs metabolised by CYP. Pantoprazole 143-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 236-239 8827397-12 1996 The effect of the acid pump inhibitors on enzymes in the liver has been intensely debated, and some authors have claimed that lansoprazole and pantoprazole have less potential than omeprazole to interact with other drugs metabolised by CYP. Omeprazole 181-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 236-239 8673608-2 1996 Cytochrome P450eryF is unusual in having alanine in place of this threonine and an ordered active site water molecule (Wat 519) which is hydrogen bonded to the substrate 5-hydroxyl group and is in position to operate as an acid catalyst required for cleaving dioxygen. Alanine 41-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8965248-6 1996 The hydroxylation of benzopyrene in human microsomes prepared from human liver samples (biopsies) was used as a model to study the treatment effect on cytochrome-P450. Benzopyrenes 21-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-166 8673608-2 1996 Cytochrome P450eryF is unusual in having alanine in place of this threonine and an ordered active site water molecule (Wat 519) which is hydrogen bonded to the substrate 5-hydroxyl group and is in position to operate as an acid catalyst required for cleaving dioxygen. Threonine 66-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8864187-0 1996 Overexpression of cytochrome P-450 isoforms involved in aflatoxin B1 bioactivation in human liver with cirrhosis and hepatitis. Aflatoxin B1 56-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 8673608-2 1996 Cytochrome P450eryF is unusual in having alanine in place of this threonine and an ordered active site water molecule (Wat 519) which is hydrogen bonded to the substrate 5-hydroxyl group and is in position to operate as an acid catalyst required for cleaving dioxygen. Water 103-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8673608-2 1996 Cytochrome P450eryF is unusual in having alanine in place of this threonine and an ordered active site water molecule (Wat 519) which is hydrogen bonded to the substrate 5-hydroxyl group and is in position to operate as an acid catalyst required for cleaving dioxygen. Hydrogen 137-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8673608-2 1996 Cytochrome P450eryF is unusual in having alanine in place of this threonine and an ordered active site water molecule (Wat 519) which is hydrogen bonded to the substrate 5-hydroxyl group and is in position to operate as an acid catalyst required for cleaving dioxygen. Oxygen 259-267 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8864187-1 1996 Studies were carried out to test the hypothesis that inflammatory liver disease increases the expression of specific cytochrome P-450 isoenzymes involved in aflatoxin B1 (AFB) activation. Aflatoxin B1 157-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 8864187-1 1996 Studies were carried out to test the hypothesis that inflammatory liver disease increases the expression of specific cytochrome P-450 isoenzymes involved in aflatoxin B1 (AFB) activation. Aflatoxin B1 171-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 8818231-0 1996 The iron(II)/reductant (DH2)-induced activation of dioxygen for the demethylation of N-methylanilines: reaction mimic for the cytochrome P-450/reductase system. ammonium ferrous sulfate 4-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-152 8818231-0 1996 The iron(II)/reductant (DH2)-induced activation of dioxygen for the demethylation of N-methylanilines: reaction mimic for the cytochrome P-450/reductase system. (-)-taxifolin 24-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-152 8818231-0 1996 The iron(II)/reductant (DH2)-induced activation of dioxygen for the demethylation of N-methylanilines: reaction mimic for the cytochrome P-450/reductase system. Oxygen 51-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-152 8818231-0 1996 The iron(II)/reductant (DH2)-induced activation of dioxygen for the demethylation of N-methylanilines: reaction mimic for the cytochrome P-450/reductase system. methylaniline 85-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-152 8875768-6 1996 In both cases, cytochrome P-450 and other enzymes that were involved in aldosterone synthesis were found mainly in tumor, but little in the zona glomerulosa of the adjacent adrenals, which showed paradoxical hyperplasia. Aldosterone 72-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 8761967-0 1996 Arachidonate metabolism in human placenta, fetal membranes, decidua and myometrium: lipoxygenase and cytochrome P450 metabolites as main products in HPLC profiles. Arachidonic Acid 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-116 8771369-2 1996 We assessed the effect of imidazole inhibitors of cytochrome P-450 on the intracytoplasmic free Ca+2 ([Ca+2]i) response generated using the microsomal ATPase inhibitor thapsigargin (THG) to deplete the intracellular Ca+2 stores. Thapsigargin 168-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 8737124-7 1996 The nitrogen atom is possibly the site that binds cytochrome P-450, which catalyses theophylline metabolism. Nitrogen 4-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 8737124-7 1996 The nitrogen atom is possibly the site that binds cytochrome P-450, which catalyses theophylline metabolism. Theophylline 84-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 8627557-9 1996 The results indicate that CYP2A6 isozyme is a major form of CYP responsible for the S-oxidation of SM-12502 in human liver microsomes. 3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one 99-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 8723743-0 1996 Characterization of the cytochrome P450 enzymes involved in the in vitro metabolism of dolasetron. dolasetron 87-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 8723743-3 1996 The compound is rapidly reduced by carbonyl reductase to form its major pharmacologically active metabolite reduced dolasetron (red-dolasetron), which us further metabolized by cytochrome P450 (CYP450). dolasetron 116-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-192 8723743-3 1996 The compound is rapidly reduced by carbonyl reductase to form its major pharmacologically active metabolite reduced dolasetron (red-dolasetron), which us further metabolized by cytochrome P450 (CYP450). dolasetron 116-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-200 8723743-4 1996 Studies were conducted, using human liver microsomes, to characterize the CYP450 enzymes responsible for the in vitro metabolism of red-dolasetron. red-dolasetron 132-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-80 8723743-7 1996 The rate of formation of 6-hydroxy red-dolasetron was significantly correlated with that of 5-hydroxy red-dolasetron, which further suggested that these metabolites were formed by the same CYP450 enzyme(s). 6-hydroxy red-dolasetron 25-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-195 8723743-7 1996 The rate of formation of 6-hydroxy red-dolasetron was significantly correlated with that of 5-hydroxy red-dolasetron, which further suggested that these metabolites were formed by the same CYP450 enzyme(s). 5-hydroxy red-dolasetron 92-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 189-195 8771369-2 1996 We assessed the effect of imidazole inhibitors of cytochrome P-450 on the intracytoplasmic free Ca+2 ([Ca+2]i) response generated using the microsomal ATPase inhibitor thapsigargin (THG) to deplete the intracellular Ca+2 stores. Thapsigargin 182-185 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 8622628-0 1996 Role of nitric oxide in the cytokine-mediated regulation of cytochrome P-450. Nitric Oxide 8-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 8622628-6 1996 The addition of inhibitors of nitric oxide synthase (NOS) significantly prevented the cytokine-mediated decrease in each CYP protein, indicating a role for nitric oxide (NO) in the down-regulation. Nitric Oxide 30-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 8622628-7 1996 Treatment of hepatocytes with the NO donor 1-hydroxy-2-oxo-3,3-bis(3-aminoethyl)-1-triazene (300 microM) caused a decrease in each CYP apoprotein, with CYP2B1/2 exhibiting the greatest decrease, to 33 +/- 8% of untreated cells. 1-hydroxy-2-oxo-3,3-bis(3-aminoethyl)-1-triazene 43-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-134 8658503-1 1996 The purpose of this study was to find out how liver injury caused by two well-known hepatotoxins, chloroform and thioacetamide, alters the expression of hepatic xenobiotic metabolizing cytochrome P450 (CYP) enzymes of DBA/2N mice. Chloroform 98-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-200 8638935-1 1996 Cytochrome P450 enzymes oxidize aldehydes either to the corresponding acid or, via a decarboxylation mechanism, to an olefin one carbon shorter than the parent substrate. Aldehydes 32-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8658503-1 1996 The purpose of this study was to find out how liver injury caused by two well-known hepatotoxins, chloroform and thioacetamide, alters the expression of hepatic xenobiotic metabolizing cytochrome P450 (CYP) enzymes of DBA/2N mice. Chloroform 98-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-205 8658503-1 1996 The purpose of this study was to find out how liver injury caused by two well-known hepatotoxins, chloroform and thioacetamide, alters the expression of hepatic xenobiotic metabolizing cytochrome P450 (CYP) enzymes of DBA/2N mice. Thioacetamide 113-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 185-200 8658503-1 1996 The purpose of this study was to find out how liver injury caused by two well-known hepatotoxins, chloroform and thioacetamide, alters the expression of hepatic xenobiotic metabolizing cytochrome P450 (CYP) enzymes of DBA/2N mice. Thioacetamide 113-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-205 8638935-1 1996 Cytochrome P450 enzymes oxidize aldehydes either to the corresponding acid or, via a decarboxylation mechanism, to an olefin one carbon shorter than the parent substrate. Alkenes 118-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8638935-1 1996 Cytochrome P450 enzymes oxidize aldehydes either to the corresponding acid or, via a decarboxylation mechanism, to an olefin one carbon shorter than the parent substrate. Carbon 129-135 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8625495-2 1996 Our previous study demonstrated that cytochrome P450 (P450) 1A2 catalyzes the formation of 4-hydroxy-1-(3-pyridyl)-1-butanone (keto alcohol) (an alpha-hydroxylation product) from NNK in human liver microsomes. 4-hydroxy-1-(3-pyridyl)-1-butanone 91-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-63 8625495-2 1996 Our previous study demonstrated that cytochrome P450 (P450) 1A2 catalyzes the formation of 4-hydroxy-1-(3-pyridyl)-1-butanone (keto alcohol) (an alpha-hydroxylation product) from NNK in human liver microsomes. keto alcohol 127-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-63 8801056-2 1996 The antifungal agent fluconazole was found to be a potent inhibitor of cytochrome P450 (P450) 2C9 (Ki = 7-8 microM), the principal enzyme responsible for the clearance (85%) of the more potent anticoagulant (S)-warfarin to the inactive (S)-7- and (S)-6-hydroxywarfarin metabolites in vivo. Fluconazole 21-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-97 8801056-2 1996 The antifungal agent fluconazole was found to be a potent inhibitor of cytochrome P450 (P450) 2C9 (Ki = 7-8 microM), the principal enzyme responsible for the clearance (85%) of the more potent anticoagulant (S)-warfarin to the inactive (S)-7- and (S)-6-hydroxywarfarin metabolites in vivo. Warfarin 211-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-97 8801056-2 1996 The antifungal agent fluconazole was found to be a potent inhibitor of cytochrome P450 (P450) 2C9 (Ki = 7-8 microM), the principal enzyme responsible for the clearance (85%) of the more potent anticoagulant (S)-warfarin to the inactive (S)-7- and (S)-6-hydroxywarfarin metabolites in vivo. ethyl-2-methylthio-4-methyl-5-pyrimidine carboxylate 236-242 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-97 8801056-1 1996 I. Inhibition of the human cytochrome P450-dependent metabolism of warfarin by fluconazole: in vitro studies. Warfarin 67-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 8801056-2 1996 The antifungal agent fluconazole was found to be a potent inhibitor of cytochrome P450 (P450) 2C9 (Ki = 7-8 microM), the principal enzyme responsible for the clearance (85%) of the more potent anticoagulant (S)-warfarin to the inactive (S)-7- and (S)-6-hydroxywarfarin metabolites in vivo. 6-hydroxywarfarin 247-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-97 8801056-1 1996 I. Inhibition of the human cytochrome P450-dependent metabolism of warfarin by fluconazole: in vitro studies. Fluconazole 79-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 8801058-3 1996 The results of studies of the effect of fluconazole on cytochrome P450 (P450) 2C9 activity in vivo and in vitro are used to develop an approach to the safe management of the warfarin-fluconazole drug interaction. Fluconazole 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-81 8801058-3 1996 The results of studies of the effect of fluconazole on cytochrome P450 (P450) 2C9 activity in vivo and in vitro are used to develop an approach to the safe management of the warfarin-fluconazole drug interaction. Warfarin 174-182 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-81 8801058-3 1996 The results of studies of the effect of fluconazole on cytochrome P450 (P450) 2C9 activity in vivo and in vitro are used to develop an approach to the safe management of the warfarin-fluconazole drug interaction. Fluconazole 183-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-81 8801060-6 1996 This route of dofetilide metabolism is mediated by cytochrome P450 (CYP). dofetilide 14-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-66 8801060-6 1996 This route of dofetilide metabolism is mediated by cytochrome P450 (CYP). dofetilide 14-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 8854046-2 1996 The study suggests that the "larger" inhibitors (such as the [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione based compounds), after an initial binding of the phenylamine nitrogen lone pair electrons with the Fe3+ haem of the cytochrome P-450, preferentially utilise the region of the AR active site which would normally bind C(17) = O of the substrate. [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione 61-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-242 8612780-1 1996 The CO-binding reaction of cytochrome P-450cam bound with (1R)-camphor and (1S)-camphor are compared in the temperature region of 210-260 K using time-resolved Fourier-transform infrared spectroscopy with the CO stretch vibration as spectroscopic probe. Camphor 58-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 8612780-1 1996 The CO-binding reaction of cytochrome P-450cam bound with (1R)-camphor and (1S)-camphor are compared in the temperature region of 210-260 K using time-resolved Fourier-transform infrared spectroscopy with the CO stretch vibration as spectroscopic probe. Camphor 75-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 8728119-0 1996 Human cytochrome P450"s are pro-oxidants in iron/ascorbate-initiated microsomal lipid peroxidation. Iron 44-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 8861658-4 1996 Dextromethorphan, debrisoquine and sparteine are good substrates for CYP2D6, whereas the S-enantiomer of mephenytoin is a good substrate for CYP2C19, both being two isozymes of cytochrome P-450. Dextromethorphan 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 8861658-4 1996 Dextromethorphan, debrisoquine and sparteine are good substrates for CYP2D6, whereas the S-enantiomer of mephenytoin is a good substrate for CYP2C19, both being two isozymes of cytochrome P-450. Debrisoquin 18-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 8861658-4 1996 Dextromethorphan, debrisoquine and sparteine are good substrates for CYP2D6, whereas the S-enantiomer of mephenytoin is a good substrate for CYP2C19, both being two isozymes of cytochrome P-450. Sparteine 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 8861658-4 1996 Dextromethorphan, debrisoquine and sparteine are good substrates for CYP2D6, whereas the S-enantiomer of mephenytoin is a good substrate for CYP2C19, both being two isozymes of cytochrome P-450. Mephenytoin 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 177-193 8647341-1 1996 Cytochrome P450, the most versatile biological catalyst known, was originally named as a pigment having a carbon monoxide difference spectrum at about 450 nm and no known function. Carbon Monoxide 106-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8728119-0 1996 Human cytochrome P450"s are pro-oxidants in iron/ascorbate-initiated microsomal lipid peroxidation. Ascorbic Acid 49-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 8854046-2 1996 The study suggests that the "larger" inhibitors (such as the [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione based compounds), after an initial binding of the phenylamine nitrogen lone pair electrons with the Fe3+ haem of the cytochrome P-450, preferentially utilise the region of the AR active site which would normally bind C(17) = O of the substrate. Aniline Compounds 159-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-242 8728119-1 1996 We have examined the effect of human cytochrome P450"s (1A1,1A2,3A4,2A6,2B6,2D6,2E1) on ascorbate/iron-induced lipid peroxidation. Ascorbic Acid 88-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 8854046-2 1996 The study suggests that the "larger" inhibitors (such as the [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione based compounds), after an initial binding of the phenylamine nitrogen lone pair electrons with the Fe3+ haem of the cytochrome P-450, preferentially utilise the region of the AR active site which would normally bind C(17) = O of the substrate. Nitrogen 171-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-242 8728119-1 1996 We have examined the effect of human cytochrome P450"s (1A1,1A2,3A4,2A6,2B6,2D6,2E1) on ascorbate/iron-induced lipid peroxidation. Iron 98-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 8728119-4 1996 It is therefore concluded that cytochrome P450"s play a significant role in ascorbate/iron peroxidation. Ascorbic Acid 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 8854046-2 1996 The study suggests that the "larger" inhibitors (such as the [2"(4"-aminophenyl)alkyl] pyrrolidine-2,5-dione based compounds), after an initial binding of the phenylamine nitrogen lone pair electrons with the Fe3+ haem of the cytochrome P-450, preferentially utilise the region of the AR active site which would normally bind C(17) = O of the substrate. fe3+ haem 209-218 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-242 8728119-4 1996 It is therefore concluded that cytochrome P450"s play a significant role in ascorbate/iron peroxidation. Iron 86-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 20650183-1 1996 Phenoxyl radicals are inevitable intermediates in the oxidative enzymatic metabolism of a phenolic antitumour drug, etoposide (VP-16), by peroxidases, cytochrome P-450, prostaglandin synthetase and tyrosinase, as well as in its interactions with oxygen and peroxyl radicals. Etoposide 116-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 8635732-0 1996 Cloning and characterization of the Saccharomyces cerevisiae C-22 sterol desaturase gene, encoding a second cytochrome P-450 involved in ergosterol biosynthesis. Ergosterol 137-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 108-124 21781659-4 1996 The pivotal role of cytochrome P450 in the mutagenicity of ochratoxin A could be demonstrated in experiments with cell lines stably expressing the human cytochrome P450 enzymes. ochratoxin A 59-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-35 21781659-4 1996 The pivotal role of cytochrome P450 in the mutagenicity of ochratoxin A could be demonstrated in experiments with cell lines stably expressing the human cytochrome P450 enzymes. ochratoxin A 59-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 153-168 21781659-10 1996 The discrepancies found between microsomal and cellular metabolism leads to the conclusion that ochratoxin A mediated mutagenicity requires additional processing of cytochrome P450 derived metabolism. ochratoxin A 96-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-180 8634277-1 1996 Two general models of the membrane topology of microsomal cytochrome P450 have been proposed: (1) deep immersion in the membrane, and (2) a P450cam-like heme domain anchored to the membrane with one or two membrane-spanning helices. Heme 153-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-73 8634277-3 1996 Cytochrome P450 2B4 was reconstituted into unilamellar phospholipid proteoliposomes (molar protein to lipid ratio 1:90). Phospholipids 55-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8907578-5 1996 In phenobarbital-treated mice, interferon beta reduced the induction of total cytochrome P-450 (22%), the activities of pentoxyresorufin O-dealkylase (38%), benzyloxyresorufin O-dealkylase (30%), erythromycin N-demethylase (30%), 7-ethoxycoumarin O-deethylase (16%) and cytochrome P-450 2B1 (33%) and 3A (45%) proteins. Phenobarbital 3-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 270-286 20650183-1 1996 Phenoxyl radicals are inevitable intermediates in the oxidative enzymatic metabolism of a phenolic antitumour drug, etoposide (VP-16), by peroxidases, cytochrome P-450, prostaglandin synthetase and tyrosinase, as well as in its interactions with oxygen and peroxyl radicals. phenoxy radical 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 8607732-2 1996 This results from macrolide inhibition of cytochrome P-450 metabolism of numerous xenobiotics, resulting in elevated serum drug levels and clinical intoxication. Macrolides 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 11859386-6 1996 This enterprise has been assisted by the determination of the amino acide sequence and the X-ray crystal structure of the bacterial cytochrome P450, P450(CAM). amino acide 62-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-147 18611706-1 1996 Itraconazole is an orally active, broad-spectrum, triazole antifungal agent which has a higher affinity for fungal cytochrome P-450 than ketoconazole but a low affinity for mammalian cytochrome P-450. Itraconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 18611706-1 1996 Itraconazole is an orally active, broad-spectrum, triazole antifungal agent which has a higher affinity for fungal cytochrome P-450 than ketoconazole but a low affinity for mammalian cytochrome P-450. Itraconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 18611706-1 1996 Itraconazole is an orally active, broad-spectrum, triazole antifungal agent which has a higher affinity for fungal cytochrome P-450 than ketoconazole but a low affinity for mammalian cytochrome P-450. Triazoles 50-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-131 8641823-5 1996 Cytochrome P-450 activity was determined at 4 and 24 hours by measuring the formation of 7-hydroxycoumarine (7-HC) from 7-ethoxycoumarine (7-EC). 7-hydroxycoumarin 89-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8555497-0 1996 Mechanisms of retinoid resistance in leukemic cells: possible role of cytochrome P450 and P-glycoprotein. Retinoids 14-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 8555497-7 1996 After isolation of microsomes, measurements showed that levels of cytochrome P450 activities in both wild-type and RA-resistant HL-60 cells were almost comparable. Tretinoin 115-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 8555497-11 1996 Possible mechanisms for accelerated clearance of RA include the induction of non-CYP1A1 cytochrome P450 enzymes and P-glycoprotein. Tretinoin 49-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 8857078-1 1996 OBJECTIVES: The biotransformation of caffeine has been studied in vitro using human cytochrome P-450 isoenzymes (CYPs) expressed in human B-lymphoblastoid cell lines, namely CYP1A1, 1A2, 2A6, 2B6, 2D6-Val, 2E1 and 3A4, and microsomal epoxide hydroxylase (EH). Caffeine 37-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 8542584-0 1996 Development of human cytochrome P450-expressing cell lines: application in mutagenicity testing of ochratoxin A. ochratoxin A 99-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 8641823-5 1996 Cytochrome P-450 activity was determined at 4 and 24 hours by measuring the formation of 7-hydroxycoumarine (7-HC) from 7-ethoxycoumarine (7-EC). 7-hydroxycoumarin 109-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8641823-5 1996 Cytochrome P-450 activity was determined at 4 and 24 hours by measuring the formation of 7-hydroxycoumarine (7-HC) from 7-ethoxycoumarine (7-EC). 7-ethoxycoumarin 120-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8641823-5 1996 Cytochrome P-450 activity was determined at 4 and 24 hours by measuring the formation of 7-hydroxycoumarine (7-HC) from 7-ethoxycoumarine (7-EC). 7-ec 139-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8533008-0 1996 Cytochrome P450: structure, function, and generation of reactive oxygen species. Reactive Oxygen Species 56-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8603034-6 1996 Cytochrome P450-dependent enzymes are involved in the synthesis and/or degradation of many endogenous compounds, such as steroids and retinoic acid. Steroids 121-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8603034-6 1996 Cytochrome P450-dependent enzymes are involved in the synthesis and/or degradation of many endogenous compounds, such as steroids and retinoic acid. Tretinoin 134-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8591087-1 1995 BACKGROUND: Liarozole binds to the cytochrome P-450-dependent hydroxylating enzymes involved in steroid biosynthesis and retinoic acid catabolism. liarozole 12-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 8896115-0 1996 Effect of imidazole derivatives on cytochrome P-450 enzyme activities in a reconstructed human epidermis. imidazole 10-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 8615873-9 1995 The apparent Km,app and Vmax,app for norfentanyl formation were 82 +/- 21 microM and 4.7 +/- 0.4 nmol product formed/min/nmol cytochrome P450, respectively. norfentanyl 37-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-141 8554320-1 1995 The NADH-dependent microsomal electron transfer system consists of NADH-cytochrome b5 reductase and cytochrome b5, which donates reducing equivalents to fatty acyl desaturase, cytochrome P450, and other reactions. NAD 4-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-191 8689938-0 1995 Metabolism of the immunosuppressant tacrolimus in the small intestine: cytochrome P450, drug interactions, and interindividual variability. Tacrolimus 36-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-86 8689938-5 1995 The cytochrome P450 (CYP) enzymes responsible for tacrolimus metabolism in small intestine were identified using specific CYP antibodies and inhibitors. Tacrolimus 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 8689938-5 1995 The cytochrome P450 (CYP) enzymes responsible for tacrolimus metabolism in small intestine were identified using specific CYP antibodies and inhibitors. Tacrolimus 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-24 8689938-5 1995 The cytochrome P450 (CYP) enzymes responsible for tacrolimus metabolism in small intestine were identified using specific CYP antibodies and inhibitors. Tacrolimus 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-125 8591087-1 1995 BACKGROUND: Liarozole binds to the cytochrome P-450-dependent hydroxylating enzymes involved in steroid biosynthesis and retinoic acid catabolism. Steroids 96-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 8624504-0 1995 The comparative study of peroxidase activity and substrate binding properties of cytochrome P450 2B4 incorporated into phospholipid vesicles with the use of two different methods. Phospholipids 119-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 8591087-1 1995 BACKGROUND: Liarozole binds to the cytochrome P-450-dependent hydroxylating enzymes involved in steroid biosynthesis and retinoic acid catabolism. Tretinoin 121-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 8591723-0 1995 Multiple forms of cytochrome P450 are involved in the metabolism of ondansetron in humans. Ondansetron 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 8750397-3 1995 Cyclosporine and tacrolimus are metabolized by the cytochrome P-450 enzyme system. Cyclosporine 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 8750397-3 1995 Cyclosporine and tacrolimus are metabolized by the cytochrome P-450 enzyme system. Tacrolimus 17-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 8591723-5 1995 The data in this study collectively indicate that multiple cytochrome P450 forms, including CYP1A1, CYP1A2, CYP2D6, and the CYP3A subfamily, are probably involved in the clearance of ondansetron in humans, with no single form of cytochrome P450 dominating the overall metabolism of ondansetron. Ondansetron 183-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 8591723-8 1995 It is therefore concluded that ondansetron is metabolized by multiple forms of cytochrome P450, and this limits the likelihood of a clinically relevant interaction with ondansetron by a modulator of a single form of cytochrome P450. Ondansetron 31-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 8591723-5 1995 The data in this study collectively indicate that multiple cytochrome P450 forms, including CYP1A1, CYP1A2, CYP2D6, and the CYP3A subfamily, are probably involved in the clearance of ondansetron in humans, with no single form of cytochrome P450 dominating the overall metabolism of ondansetron. Ondansetron 183-194 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 229-244 8591723-8 1995 It is therefore concluded that ondansetron is metabolized by multiple forms of cytochrome P450, and this limits the likelihood of a clinically relevant interaction with ondansetron by a modulator of a single form of cytochrome P450. Ondansetron 31-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-231 8591723-8 1995 It is therefore concluded that ondansetron is metabolized by multiple forms of cytochrome P450, and this limits the likelihood of a clinically relevant interaction with ondansetron by a modulator of a single form of cytochrome P450. Ondansetron 169-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-94 8591723-5 1995 The data in this study collectively indicate that multiple cytochrome P450 forms, including CYP1A1, CYP1A2, CYP2D6, and the CYP3A subfamily, are probably involved in the clearance of ondansetron in humans, with no single form of cytochrome P450 dominating the overall metabolism of ondansetron. Ondansetron 282-293 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-74 8591723-8 1995 It is therefore concluded that ondansetron is metabolized by multiple forms of cytochrome P450, and this limits the likelihood of a clinically relevant interaction with ondansetron by a modulator of a single form of cytochrome P450. Ondansetron 169-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 216-231 8588954-7 1995 Isolated HPTC also maintained levels of reduced glutathione (GSH) (11.9 +/- 3.2 nmol mg-1 protein) and exhibited cytochrome P450-dependent activity, levels of spectrally determined P450 being 0.22 +/- 0.07 nmol mg-1 protein. hptc 9-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 8591727-1 1995 The aim of this study was to identify the form(s) of cytochrome P450 (CYP) responsible for the biotransformation of zolpidem to its alcohol derivatives which, after rapid conversion to carboxylic acids, represents the main way of metabolism in humans. Zolpidem 116-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 8591727-1 1995 The aim of this study was to identify the form(s) of cytochrome P450 (CYP) responsible for the biotransformation of zolpidem to its alcohol derivatives which, after rapid conversion to carboxylic acids, represents the main way of metabolism in humans. Zolpidem 116-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 8591727-1 1995 The aim of this study was to identify the form(s) of cytochrome P450 (CYP) responsible for the biotransformation of zolpidem to its alcohol derivatives which, after rapid conversion to carboxylic acids, represents the main way of metabolism in humans. Alcohols 132-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 8591727-1 1995 The aim of this study was to identify the form(s) of cytochrome P450 (CYP) responsible for the biotransformation of zolpidem to its alcohol derivatives which, after rapid conversion to carboxylic acids, represents the main way of metabolism in humans. Alcohols 132-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 8591727-1 1995 The aim of this study was to identify the form(s) of cytochrome P450 (CYP) responsible for the biotransformation of zolpidem to its alcohol derivatives which, after rapid conversion to carboxylic acids, represents the main way of metabolism in humans. Carboxylic Acids 185-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 8591727-1 1995 The aim of this study was to identify the form(s) of cytochrome P450 (CYP) responsible for the biotransformation of zolpidem to its alcohol derivatives which, after rapid conversion to carboxylic acids, represents the main way of metabolism in humans. Carboxylic Acids 185-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-73 8690232-2 1995 Dapsone is a potent anti-inflammatory and anti-parasitic compound, which is metabolised by cytochrome P-450 to hydroxylamines, which in turn cause methaemoglobinaemia and haemolysis. Dapsone 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 8690232-2 1995 Dapsone is a potent anti-inflammatory and anti-parasitic compound, which is metabolised by cytochrome P-450 to hydroxylamines, which in turn cause methaemoglobinaemia and haemolysis. Hydroxylamines 111-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 7559586-0 1995 The stoichiometry of the cytochrome P-450-catalyzed metabolism of methoxyflurane and benzphetamine in the presence and absence of cytochrome b5. Methoxyflurane 66-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 7559586-0 1995 The stoichiometry of the cytochrome P-450-catalyzed metabolism of methoxyflurane and benzphetamine in the presence and absence of cytochrome b5. Benzphetamine 85-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 8654202-0 1995 Identification of human cytochrome P450 isozymes responsible for the in vitro oxidative metabolism of finasteride. Finasteride 102-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 7559586-1 1995 The complete stoichiometry of the metabolism of the cytochrome b5 (cyt b5)-requiring substrate, methoxyflurane, by purified cytochrome P-450 2B4 was compared to that of another substrate, benzphetamine, which does not require cyt b5 for its metabolism. Methoxyflurane 96-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 7586124-3 1995 Oxidation of butadiene to 1,2-epoxy-3-butene (BMO) and further activation to BDE is catalysed by cytochrome P450 (CYP) isozymes. 1,3-butadiene 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-112 7586124-3 1995 Oxidation of butadiene to 1,2-epoxy-3-butene (BMO) and further activation to BDE is catalysed by cytochrome P450 (CYP) isozymes. 1,3-butadiene 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 7586124-3 1995 Oxidation of butadiene to 1,2-epoxy-3-butene (BMO) and further activation to BDE is catalysed by cytochrome P450 (CYP) isozymes. 3,4-epoxy-1-butene 26-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-112 7586124-3 1995 Oxidation of butadiene to 1,2-epoxy-3-butene (BMO) and further activation to BDE is catalysed by cytochrome P450 (CYP) isozymes. 3,4-epoxy-1-butene 26-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 7586124-3 1995 Oxidation of butadiene to 1,2-epoxy-3-butene (BMO) and further activation to BDE is catalysed by cytochrome P450 (CYP) isozymes. bmo 46-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-112 7586124-3 1995 Oxidation of butadiene to 1,2-epoxy-3-butene (BMO) and further activation to BDE is catalysed by cytochrome P450 (CYP) isozymes. bmo 46-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-117 8654206-0 1995 Identification of the human liver cytochrome P450 enzymes involved in the metabolism of zileuton (ABT-077) and its N-dehydroxylated metabolite, Abbott-66193. zileuton 88-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 8654206-0 1995 Identification of the human liver cytochrome P450 enzymes involved in the metabolism of zileuton (ABT-077) and its N-dehydroxylated metabolite, Abbott-66193. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 98-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 8654206-1 1995 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]zileuton (ABT-077) and its N-dehydroxylated metabolite, [14C]Abbott-66193, by human liver microsomes. Carbon-14 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 8654206-1 1995 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]zileuton (ABT-077) and its N-dehydroxylated metabolite, [14C]Abbott-66193, by human liver microsomes. Carbon-14 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 8654206-1 1995 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]zileuton (ABT-077) and its N-dehydroxylated metabolite, [14C]Abbott-66193, by human liver microsomes. zileuton 129-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 8654206-1 1995 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]zileuton (ABT-077) and its N-dehydroxylated metabolite, [14C]Abbott-66193, by human liver microsomes. zileuton 129-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 8654206-1 1995 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]zileuton (ABT-077) and its N-dehydroxylated metabolite, [14C]Abbott-66193, by human liver microsomes. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 139-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 7588807-0 1995 Compressibility of the heme pocket of substrate analogue complexes of cytochrome P-450cam-CO. Heme 23-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-92 16535142-3 1995 The enzyme responsible for this demethylation (31-O-FK-506 demethylase) was isolated and shown to be a soluble cytoplasmic protein which is constitutively expressed in the cells, which requires NADPH, ferredoxin-NADP(sup+)-reductase, and ferredoxin for activity, and which shows a cytochrome P-450 light absorption characteristic. NADP 194-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 281-297 16535142-4 1995 Carbon monoxide saturation of the enzyme preparation and known mammalian cytochrome P-450 inhibitors such as quinidine HCl, ketoconazole, troleandomycin, and sulfaphenazole abolish the demethylating activity extensively. QUINIDINE HYDROCHLORIDE 109-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 16535142-4 1995 Carbon monoxide saturation of the enzyme preparation and known mammalian cytochrome P-450 inhibitors such as quinidine HCl, ketoconazole, troleandomycin, and sulfaphenazole abolish the demethylating activity extensively. Ketoconazole 124-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 16535142-4 1995 Carbon monoxide saturation of the enzyme preparation and known mammalian cytochrome P-450 inhibitors such as quinidine HCl, ketoconazole, troleandomycin, and sulfaphenazole abolish the demethylating activity extensively. Troleandomycin 138-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 16535142-4 1995 Carbon monoxide saturation of the enzyme preparation and known mammalian cytochrome P-450 inhibitors such as quinidine HCl, ketoconazole, troleandomycin, and sulfaphenazole abolish the demethylating activity extensively. Sulfaphenazole 158-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 16535142-6 1995 The isolated demethylase is therefore a cytochrome P-450 protein that can be used as a catalyst for the synthesis of 31-O-desmethylFK-506, an important immunosuppressant and a known metabolite of FK-506 metabolism by human liver microsomes. 31-o- 117-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 16535142-6 1995 The isolated demethylase is therefore a cytochrome P-450 protein that can be used as a catalyst for the synthesis of 31-O-desmethylFK-506, an important immunosuppressant and a known metabolite of FK-506 metabolism by human liver microsomes. Tacrolimus 131-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 8654192-7 1995 The involvement of cytochrome P450 (CYP) in the metabolism of mofarotene was examined with human liver microsomes. mofarotene 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-34 8654192-7 1995 The involvement of cytochrome P450 (CYP) in the metabolism of mofarotene was examined with human liver microsomes. mofarotene 62-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 8654206-1 1995 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]zileuton (ABT-077) and its N-dehydroxylated metabolite, [14C]Abbott-66193, by human liver microsomes. 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 139-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 8654206-1 1995 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]zileuton (ABT-077) and its N-dehydroxylated metabolite, [14C]Abbott-66193, by human liver microsomes. Carbon-14 186-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 8654206-1 1995 In vitro studies were conducted to identify the hepatic cytochrome P450 (CYP) forms involved in the oxidative metabolism of [14C]zileuton (ABT-077) and its N-dehydroxylated metabolite, [14C]Abbott-66193, by human liver microsomes. Carbon-14 186-189 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 8654206-3 1995 Results suggested that whereas the metabolism of zileuton and Abbott-66193 were mediated by the same CYP forms, the CYP forms responsible for hydroxylation (CYP1A2 and CYP2C9/10) were distinct from those involved in sulfoxidation (CYP3A > CYP2C9/10). zileuton 49-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-104 8654202-2 1995 In this study, we identified specific human cytochrome P450 (CYP) isozyme(s) involved in the in vitro metabolism of [14C]finasteride using CYP isozyme-selective inhibitors and microsomes containing specific recombinant human CYP isozymes (expressed in human AHH-1 TK+/-cells). [14c]finasteride 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-59 8654202-2 1995 In this study, we identified specific human cytochrome P450 (CYP) isozyme(s) involved in the in vitro metabolism of [14C]finasteride using CYP isozyme-selective inhibitors and microsomes containing specific recombinant human CYP isozymes (expressed in human AHH-1 TK+/-cells). [14c]finasteride 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-64 8654202-2 1995 In this study, we identified specific human cytochrome P450 (CYP) isozyme(s) involved in the in vitro metabolism of [14C]finasteride using CYP isozyme-selective inhibitors and microsomes containing specific recombinant human CYP isozymes (expressed in human AHH-1 TK+/-cells). [14c]finasteride 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 8654202-2 1995 In this study, we identified specific human cytochrome P450 (CYP) isozyme(s) involved in the in vitro metabolism of [14C]finasteride using CYP isozyme-selective inhibitors and microsomes containing specific recombinant human CYP isozymes (expressed in human AHH-1 TK+/-cells). [14c]finasteride 116-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-142 8654202-9 1995 Other selective CYP inhibitors for CYP1A1/2 (alpha-naphthoflavone), CYP2C8-10 (sulfaphenazole), CYP2D6 (quinidine), and CYP2E1 (diallylsulfone) showed minor or no effects on both reactions. alpha-naphthoflavone 45-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 8654202-9 1995 Other selective CYP inhibitors for CYP1A1/2 (alpha-naphthoflavone), CYP2C8-10 (sulfaphenazole), CYP2D6 (quinidine), and CYP2E1 (diallylsulfone) showed minor or no effects on both reactions. Sulfaphenazole 79-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 8654202-9 1995 Other selective CYP inhibitors for CYP1A1/2 (alpha-naphthoflavone), CYP2C8-10 (sulfaphenazole), CYP2D6 (quinidine), and CYP2E1 (diallylsulfone) showed minor or no effects on both reactions. Quinidine 104-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 8654202-9 1995 Other selective CYP inhibitors for CYP1A1/2 (alpha-naphthoflavone), CYP2C8-10 (sulfaphenazole), CYP2D6 (quinidine), and CYP2E1 (diallylsulfone) showed minor or no effects on both reactions. diallyl sulfone 128-142 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-19 7496999-11 1995 The greatest increase in N-demethylation activity was observed in the reconstitution system with the lowest concentration of cytochrome P-450 reductase, conditions which most closely resemble intact microsomes. Nitrogen 25-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 8847710-0 1995 Some aspects of the role of cytochrome P-450 isozymes in the N-oxidative transformation of secondary and tertiary amine compounds. Amines 114-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 8847710-1 1995 Indirect evidence of the participation of cytochrome P-450 (P-450) in the microsomal N-oxygenation of secondary and tertiary nitrogen functions is presented by studies employing diagnostic modifiers of the hemoprotein system as well as antibodies directed toward the diverse P-450 isoforms and NADPH-cytochrome P-450 reductase. Nitrogen 85-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 8847710-1 1995 Indirect evidence of the participation of cytochrome P-450 (P-450) in the microsomal N-oxygenation of secondary and tertiary nitrogen functions is presented by studies employing diagnostic modifiers of the hemoprotein system as well as antibodies directed toward the diverse P-450 isoforms and NADPH-cytochrome P-450 reductase. Nitrogen 125-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 7636727-2 1995 We report the effect of a known cytochrome P450 (CYP) inducer, rifampicin, on the metabolism of lidocaine by primary human hepatocytes. Rifampin 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 7491124-5 1995 The alterations, by beta-carbolines, of some important enzymatic systems, e.g. cytochrome P-450, have been clearly demonstrated, yet many discrepancies and contradictions exist so that an interpretation of the results and the definition of some common mechanism appears premature. Carbolines 20-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 79-95 7493150-1 1995 The cytochrome P450-dependent monooxygenases constitute the primary enzyme system responsible for the oxidative metabolism of a variety of xenobiotics and endogenous compounds including drugs, carcinogens, fatty acids and hormones. Fatty Acids 206-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 7636727-2 1995 We report the effect of a known cytochrome P450 (CYP) inducer, rifampicin, on the metabolism of lidocaine by primary human hepatocytes. Rifampin 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 7636727-2 1995 We report the effect of a known cytochrome P450 (CYP) inducer, rifampicin, on the metabolism of lidocaine by primary human hepatocytes. Lidocaine 96-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-47 7636727-2 1995 We report the effect of a known cytochrome P450 (CYP) inducer, rifampicin, on the metabolism of lidocaine by primary human hepatocytes. Lidocaine 96-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 7636727-3 1995 Rifampicin has been shown to induce CYP3A4, a major human hepatic CYP isozyme that is known to metabolize lidocaine to its primary metabolite, monoethylglycinexylidide. Rifampin 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 7636727-3 1995 Rifampicin has been shown to induce CYP3A4, a major human hepatic CYP isozyme that is known to metabolize lidocaine to its primary metabolite, monoethylglycinexylidide. Lidocaine 106-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 7636727-3 1995 Rifampicin has been shown to induce CYP3A4, a major human hepatic CYP isozyme that is known to metabolize lidocaine to its primary metabolite, monoethylglycinexylidide. monoethylglycinexylidide 143-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 8536426-2 1995 The high probability of interactions can be explained by two pharmacokinetic properties of coumarins: high binding to plasma albumin (99%), being displaced by other drugs with greater affinity to this protein, and metabolism by liver microsomal enzymes (cytochrome P450), which can be induced or inhibited by other compounds (Shinn & Shrewsbury 1985). Coumarins 91-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 254-269 7623862-2 1995 BD is metabolized by cytochrome P-450-dependent monoxygenases to the primary metabolite 1,2-epoxybutene-3 (epoxybutene, EB). 3,4-epoxy-1-butene 88-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 7623862-2 1995 BD is metabolized by cytochrome P-450-dependent monoxygenases to the primary metabolite 1,2-epoxybutene-3 (epoxybutene, EB). 3,4-epoxy-1-butene 92-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 7623862-2 1995 BD is metabolized by cytochrome P-450-dependent monoxygenases to the primary metabolite 1,2-epoxybutene-3 (epoxybutene, EB). ethylbenzene 120-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 7622489-0 1995 Different mechanisms of hydroxylation site selection by liver and kidney cytochrome P450 species (CYP27 and CYP24) involved in vitamin D metabolism. Vitamin D 127-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 7622489-3 1995 Lengthening the side chain is tolerated by each cytochrome P450 isoform such that 25-hydroxylation or 24-hydroxylation continues to occur at the same rate as in the native side chain, while the site of hydroxylation remains the same for the liver enzyme in that CYP27 continues to hydroxylate at C-25 and C-27 (minor) despite the two-carbon-atom extension. Carbon 262-263 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 7622489-3 1995 Lengthening the side chain is tolerated by each cytochrome P450 isoform such that 25-hydroxylation or 24-hydroxylation continues to occur at the same rate as in the native side chain, while the site of hydroxylation remains the same for the liver enzyme in that CYP27 continues to hydroxylate at C-25 and C-27 (minor) despite the two-carbon-atom extension. Carbon 296-297 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 7622489-3 1995 Lengthening the side chain is tolerated by each cytochrome P450 isoform such that 25-hydroxylation or 24-hydroxylation continues to occur at the same rate as in the native side chain, while the site of hydroxylation remains the same for the liver enzyme in that CYP27 continues to hydroxylate at C-25 and C-27 (minor) despite the two-carbon-atom extension. Carbon 334-340 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-63 7625847-1 1995 The membrane-bound endogenous fatty acid arachidonic acid can be released from membranes by phospholipases and then metabolized to biologically active compounds by cyclooxygenases, lipoxygenases, and cytochrome P450 (CYP) enzymes. Fatty Acids 30-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-215 7625847-1 1995 The membrane-bound endogenous fatty acid arachidonic acid can be released from membranes by phospholipases and then metabolized to biologically active compounds by cyclooxygenases, lipoxygenases, and cytochrome P450 (CYP) enzymes. Fatty Acids 30-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 217-220 7625847-1 1995 The membrane-bound endogenous fatty acid arachidonic acid can be released from membranes by phospholipases and then metabolized to biologically active compounds by cyclooxygenases, lipoxygenases, and cytochrome P450 (CYP) enzymes. Arachidonic Acid 41-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-215 7625847-1 1995 The membrane-bound endogenous fatty acid arachidonic acid can be released from membranes by phospholipases and then metabolized to biologically active compounds by cyclooxygenases, lipoxygenases, and cytochrome P450 (CYP) enzymes. Arachidonic Acid 41-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 217-220 7625847-3 1995 Liver CYP arachidonate products include epoxyeicosatrienoic acids (EETs) and monohydroxylated products (HETEs). Arachidonic Acid 10-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 7625847-3 1995 Liver CYP arachidonate products include epoxyeicosatrienoic acids (EETs) and monohydroxylated products (HETEs). epoxyeicosatrienoic acids 40-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 7625847-3 1995 Liver CYP arachidonate products include epoxyeicosatrienoic acids (EETs) and monohydroxylated products (HETEs). eets 67-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-9 7625847-4 1995 We examined metabolism of [1-14C]arachidonic acid by a panel of 10 human CYP enzymes expressed in HepG2 cells. Arachidonic Acid 26-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-76 7788513-13 1995 Cross-reactivity with other anticonvulsant agents capable of forming arene oxide intermediates occurs in the cytochrome P-450 system. Benzene oxide 69-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 8536426-2 1995 The high probability of interactions can be explained by two pharmacokinetic properties of coumarins: high binding to plasma albumin (99%), being displaced by other drugs with greater affinity to this protein, and metabolism by liver microsomal enzymes (cytochrome P450), which can be induced or inhibited by other compounds (Shinn & Shrewsbury 1985). shinn & 326-336 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 254-269 8586256-0 1995 Probing the cytochrome P-450 2B1 active site with diamantoid compounds. diamantoid 50-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 8586256-1 1995 Hydrocarbone diamantane has been shown to be a specific substrate with a high affinity for the binding site of PB-inducible cytochrome P-450 2B1 (Hodek et al. hydrocarbone diamantane 0-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 8586256-7 1995 Of all oxygen containing derivatives diamantane 1,6-dicarboxylic acid dimethylester only exhibited a pronounced ligand interaction with cytochrome P-450. diamantane 1,6-dicarboxylic acid dimethylester 37-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-152 8586256-1 1995 Hydrocarbone diamantane has been shown to be a specific substrate with a high affinity for the binding site of PB-inducible cytochrome P-450 2B1 (Hodek et al. Lead 111-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-140 8586256-4 1995 Of the compounds (diamantane and its analogues, adamantane and triamantane) tested, diamantane had the lowest value of a spectral dissociation constant Ks = 0.5 mumol/l, indicating that diamantane was accommodated well to the cytochrome P-450 2B1, hence values of 0.46 nm and 0.66 nm for the width and length of the diamantane molecule, respectively, were used to describe of the dimensions the cytochrome P-450 binding site. Adamantane 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-242 8586256-4 1995 Of the compounds (diamantane and its analogues, adamantane and triamantane) tested, diamantane had the lowest value of a spectral dissociation constant Ks = 0.5 mumol/l, indicating that diamantane was accommodated well to the cytochrome P-450 2B1, hence values of 0.46 nm and 0.66 nm for the width and length of the diamantane molecule, respectively, were used to describe of the dimensions the cytochrome P-450 binding site. Adamantane 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 395-411 8586256-4 1995 Of the compounds (diamantane and its analogues, adamantane and triamantane) tested, diamantane had the lowest value of a spectral dissociation constant Ks = 0.5 mumol/l, indicating that diamantane was accommodated well to the cytochrome P-450 2B1, hence values of 0.46 nm and 0.66 nm for the width and length of the diamantane molecule, respectively, were used to describe of the dimensions the cytochrome P-450 binding site. Adamantane 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-242 8586256-4 1995 Of the compounds (diamantane and its analogues, adamantane and triamantane) tested, diamantane had the lowest value of a spectral dissociation constant Ks = 0.5 mumol/l, indicating that diamantane was accommodated well to the cytochrome P-450 2B1, hence values of 0.46 nm and 0.66 nm for the width and length of the diamantane molecule, respectively, were used to describe of the dimensions the cytochrome P-450 binding site. Adamantane 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 395-411 8586256-4 1995 Of the compounds (diamantane and its analogues, adamantane and triamantane) tested, diamantane had the lowest value of a spectral dissociation constant Ks = 0.5 mumol/l, indicating that diamantane was accommodated well to the cytochrome P-450 2B1, hence values of 0.46 nm and 0.66 nm for the width and length of the diamantane molecule, respectively, were used to describe of the dimensions the cytochrome P-450 binding site. Adamantane 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-242 8586256-4 1995 Of the compounds (diamantane and its analogues, adamantane and triamantane) tested, diamantane had the lowest value of a spectral dissociation constant Ks = 0.5 mumol/l, indicating that diamantane was accommodated well to the cytochrome P-450 2B1, hence values of 0.46 nm and 0.66 nm for the width and length of the diamantane molecule, respectively, were used to describe of the dimensions the cytochrome P-450 binding site. Adamantane 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 395-411 8586256-7 1995 Of all oxygen containing derivatives diamantane 1,6-dicarboxylic acid dimethylester only exhibited a pronounced ligand interaction with cytochrome P-450. Oxygen 7-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-152 8685072-12 1995 This study suggests, that fluoxetine and fluvoxamine differ in their interaction with the metabolism of some other basic psychotropic drugs, by a mechanism which implies CYP2D6 and CYPmeph and possibly other isoformes of cytochrome P-450. Fluoxetine 26-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-237 8685072-12 1995 This study suggests, that fluoxetine and fluvoxamine differ in their interaction with the metabolism of some other basic psychotropic drugs, by a mechanism which implies CYP2D6 and CYPmeph and possibly other isoformes of cytochrome P-450. Fluvoxamine 41-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-237 7750560-1 1995 13CO NMR chemical shifts and 12CO infrared stretching frequencies have been measured for cytochrome P-450cam-CO in the presence of D-camphor and of various camphor analogues. Camphor 131-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-111 7539291-8 1995 Not only does the heme pocket of NOS have the same coordination as cytochrome P-450 in its stable form, but the partially denatured form has the same properties as cytochrome P-420, the inactive form of cytochrome P-450. Heme 18-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 203-219 7750560-1 1995 13CO NMR chemical shifts and 12CO infrared stretching frequencies have been measured for cytochrome P-450cam-CO in the presence of D-camphor and of various camphor analogues. Camphor 133-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-111 7723077-9 1995 We conclude that DES is metabolized to histone-binding metabolites, presumably by nuclear cytochrome P-450. Diethylstilbestrol 17-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 7640150-0 1995 Identification of human CYP isoforms involved in the metabolism of propranolol enantiomers--N-desisopropylation is mediated mainly by CYP1A2. Propranolol 67-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-27 7640150-2 1995 Studies using human liver microsomes and six recombinant human CYP isoforms (i.e. CYP1A2, 2A6, 2B6, 2D6, 2E1 and 3A4) were performed to identify the cytochrome P450 (CYP) isoform(s) involved in the ring 4-hydroxylation and side-chain N-desisopropylation of propranolol enantiomers in humans. Nitrogen 234-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-164 7640150-2 1995 Studies using human liver microsomes and six recombinant human CYP isoforms (i.e. CYP1A2, 2A6, 2B6, 2D6, 2E1 and 3A4) were performed to identify the cytochrome P450 (CYP) isoform(s) involved in the ring 4-hydroxylation and side-chain N-desisopropylation of propranolol enantiomers in humans. Propranolol 257-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-164 7627274-7 1995 Immunohistochemical analysis revealed that tumor cells were positively stained for C21 hydroxylase cytochrome P-450 (P-450C21) and P-450(11) beta which convert 17-hydroxy (OH) progesterone to cortisol as well as P-450SCC, 3 beta-hydroxysteroid dehydrogenase and P-450(17) alpha which are involved in testosterone biosynthesis. 17-hydroxy (oh) progesterone 160-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 7627274-7 1995 Immunohistochemical analysis revealed that tumor cells were positively stained for C21 hydroxylase cytochrome P-450 (P-450C21) and P-450(11) beta which convert 17-hydroxy (OH) progesterone to cortisol as well as P-450SCC, 3 beta-hydroxysteroid dehydrogenase and P-450(17) alpha which are involved in testosterone biosynthesis. Hydrocortisone 192-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 7627274-7 1995 Immunohistochemical analysis revealed that tumor cells were positively stained for C21 hydroxylase cytochrome P-450 (P-450C21) and P-450(11) beta which convert 17-hydroxy (OH) progesterone to cortisol as well as P-450SCC, 3 beta-hydroxysteroid dehydrogenase and P-450(17) alpha which are involved in testosterone biosynthesis. Testosterone 300-312 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 7796864-7 1995 Dexamethasone, already used in patients to reduce the toxicity of interleukin-2 therapy, provided full protection against the cytochrome P-450 depression. Dexamethasone 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-142 7663534-0 1995 Oxidative metabolism of bunitrolol by complementary DNA-expressed human cytochrome P450 isozymes in a human hepatoma cell line (Hep G2) using recombinant vaccinia virus. bunitrolol 24-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 7620235-12 1995 This again was to be expected, considering the known inhibitory effect of cimetidine on cytochrome P450. Cimetidine 74-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-103 7616365-1 1995 This study was conducted in eight healthy volunteers to assess the time course of induction of cytochrome P-450 by phenytoin. Phenytoin 115-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 7628308-0 1995 N4-hydroxylation of sulfamethoxazole by cytochrome P450 of the cytochrome P4502C subfamily and reduction of sulfamethoxazole hydroxylamine in human and rat hepatic microsomes. Sulfamethoxazole 20-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 7534104-3 1995 Several studies indicate that cytochrome P450-dependent metabolites of sulphonamides act as the nominal allergens. Sulfonamides 71-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 7537544-0 1995 Role of cytochrome P-450 enzymes and metabolites of arachidonic acid in the control of vascular tone. Arachidonic Acid 52-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 7864832-20 1995 The striking contrast between DBM and cytochrome P-450, which carries out both epoxidation and allylic oxidation with non-conjugated olefinic substrates, is probably a reflection of the differences in redox potential of the activated oxygen species operative for these two enzymes. olefinic 133-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 7864832-20 1995 The striking contrast between DBM and cytochrome P-450, which carries out both epoxidation and allylic oxidation with non-conjugated olefinic substrates, is probably a reflection of the differences in redox potential of the activated oxygen species operative for these two enzymes. Oxygen 234-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 7891318-2 1995 Kinetic and inhibitor studies with human liver microsomes and complementary DNA-expressed enzyme were performed to identify the cytochrome P450 (CYP) isoform responsible for torsemide tolyl methylhydroxylation to predict factors that might alter clearance in patients receiving torsemide. Torsemide 174-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-143 7891318-2 1995 Kinetic and inhibitor studies with human liver microsomes and complementary DNA-expressed enzyme were performed to identify the cytochrome P450 (CYP) isoform responsible for torsemide tolyl methylhydroxylation to predict factors that might alter clearance in patients receiving torsemide. Torsemide 174-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 7891318-2 1995 Kinetic and inhibitor studies with human liver microsomes and complementary DNA-expressed enzyme were performed to identify the cytochrome P450 (CYP) isoform responsible for torsemide tolyl methylhydroxylation to predict factors that might alter clearance in patients receiving torsemide. Torsemide 278-287 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-143 7891318-2 1995 Kinetic and inhibitor studies with human liver microsomes and complementary DNA-expressed enzyme were performed to identify the cytochrome P450 (CYP) isoform responsible for torsemide tolyl methylhydroxylation to predict factors that might alter clearance in patients receiving torsemide. Torsemide 278-287 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 7850793-6 1995 Plasma concentrations of dechlorethylcyclophosphamide and carboxyphosphamide were correlated in individual patients, suggesting that the activity of both aldehyde dehydrogenase and cytochrome P450 enzyme(s) determine carboxyphosphamide production in vivo. dechlorethylcyclophosphamide 25-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-196 7850793-6 1995 Plasma concentrations of dechlorethylcyclophosphamide and carboxyphosphamide were correlated in individual patients, suggesting that the activity of both aldehyde dehydrogenase and cytochrome P450 enzyme(s) determine carboxyphosphamide production in vivo. carboxyphosphamide 58-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-196 7850793-6 1995 Plasma concentrations of dechlorethylcyclophosphamide and carboxyphosphamide were correlated in individual patients, suggesting that the activity of both aldehyde dehydrogenase and cytochrome P450 enzyme(s) determine carboxyphosphamide production in vivo. carboxyphosphamide 217-235 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-196 7736913-5 1995 The oxidation of both losartan and the putative aldehyde intermediate E3179 was catalyzed by the microsomal fraction, required both NADPH and molecular oxygen, and was inhibited by SKF 525-A, implicating cytochrome P450 (CYP). Losartan 22-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-219 7540767-3 1995 In order for AFB1 to exert its effects, it must be converted to its reactive epoxide by the action of the mixed function mono-oxygenase enzyme systems (cytochrome P450-dependent) in the tissues (in particular, the liver) of the affected animal. Aflatoxin B1 13-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-167 7540767-3 1995 In order for AFB1 to exert its effects, it must be converted to its reactive epoxide by the action of the mixed function mono-oxygenase enzyme systems (cytochrome P450-dependent) in the tissues (in particular, the liver) of the affected animal. Epoxy Compounds 77-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-167 7736913-5 1995 The oxidation of both losartan and the putative aldehyde intermediate E3179 was catalyzed by the microsomal fraction, required both NADPH and molecular oxygen, and was inhibited by SKF 525-A, implicating cytochrome P450 (CYP). Losartan 22-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-224 7736913-5 1995 The oxidation of both losartan and the putative aldehyde intermediate E3179 was catalyzed by the microsomal fraction, required both NADPH and molecular oxygen, and was inhibited by SKF 525-A, implicating cytochrome P450 (CYP). Aldehydes 48-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-219 7736913-5 1995 The oxidation of both losartan and the putative aldehyde intermediate E3179 was catalyzed by the microsomal fraction, required both NADPH and molecular oxygen, and was inhibited by SKF 525-A, implicating cytochrome P450 (CYP). Aldehydes 48-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-224 7736913-5 1995 The oxidation of both losartan and the putative aldehyde intermediate E3179 was catalyzed by the microsomal fraction, required both NADPH and molecular oxygen, and was inhibited by SKF 525-A, implicating cytochrome P450 (CYP). e3179 70-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-219 7736913-5 1995 The oxidation of both losartan and the putative aldehyde intermediate E3179 was catalyzed by the microsomal fraction, required both NADPH and molecular oxygen, and was inhibited by SKF 525-A, implicating cytochrome P450 (CYP). e3179 70-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-224 7736913-5 1995 The oxidation of both losartan and the putative aldehyde intermediate E3179 was catalyzed by the microsomal fraction, required both NADPH and molecular oxygen, and was inhibited by SKF 525-A, implicating cytochrome P450 (CYP). Proadifen 181-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 204-219 7840155-1 1995 Cytochrome P-450 (P-450) is a NADPH-requiring and O2-dependent monooxygenase system. NADP 30-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 15299332-0 1995 Modeling protein-substrate interactions in the heme domain of cytochrome P450(BM-3). Heme 47-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-82 7840218-1 1995 Leukotoxin (Lx), a cytochrome P-450-dependent metabolite of linoleate synthesized by neutrophils or synthesized by OH- and linoleate in neutrophil cell membranes, has been recovered in lung lavages of patients with the adult respiratory distress syndrome. Linoleic Acid 60-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 7967726-8 1994 Both of these hypercatabolic states can be modulated by concurrent administration of ketoconazole, an inhibitor of cytochrome P-450 and lipoxygenase-mediated oxidations. Ketoconazole 85-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-148 7703274-1 1995 The NADPH-adrenodoxin complex with adrenodoxin is responsible for the transformation of the two-electron flow from NADH to the mono-electron flow to cytochrome P-450 in the steroid-hydroxyl enzyme system of mitochondria of kidney crust. nadph-adrenodoxin 4-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 7703274-1 1995 The NADPH-adrenodoxin complex with adrenodoxin is responsible for the transformation of the two-electron flow from NADH to the mono-electron flow to cytochrome P-450 in the steroid-hydroxyl enzyme system of mitochondria of kidney crust. NAD 115-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 7703274-1 1995 The NADPH-adrenodoxin complex with adrenodoxin is responsible for the transformation of the two-electron flow from NADH to the mono-electron flow to cytochrome P-450 in the steroid-hydroxyl enzyme system of mitochondria of kidney crust. Steroids 173-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Fluvoxamine 335-346 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Venlafaxine Hydrochloride 352-363 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Venlafaxine Hydrochloride 352-363 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 7756927-6 1995 This mechanism operates with all the substrates and all cytochrome P-450 forms capable of catalyzing the partially coupled monooxygenase reactions proceeding with the formation of hydrogen peroxide as a by-product. Hydrogen Peroxide 180-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 7981206-1 1994 Camphor binding to ferric cytochrome P-450cam is a two-step process. Camphor 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 7704038-1 1994 Metabolism of diazepam was studied in vitro to identify the forms of cytochrome P450 (CYP) responsible for N-demethylation (nordazepam formation) and 3-hydroxylation (temazepam formation), using liver microsomes obtained from extensive (EM) and poor metabolizers (PM) for S-mephenytoin 4"-hydroxylation. Diazepam 14-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 7704038-1 1994 Metabolism of diazepam was studied in vitro to identify the forms of cytochrome P450 (CYP) responsible for N-demethylation (nordazepam formation) and 3-hydroxylation (temazepam formation), using liver microsomes obtained from extensive (EM) and poor metabolizers (PM) for S-mephenytoin 4"-hydroxylation. Diazepam 14-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 7704038-1 1994 Metabolism of diazepam was studied in vitro to identify the forms of cytochrome P450 (CYP) responsible for N-demethylation (nordazepam formation) and 3-hydroxylation (temazepam formation), using liver microsomes obtained from extensive (EM) and poor metabolizers (PM) for S-mephenytoin 4"-hydroxylation. Nordazepam 124-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 7704038-1 1994 Metabolism of diazepam was studied in vitro to identify the forms of cytochrome P450 (CYP) responsible for N-demethylation (nordazepam formation) and 3-hydroxylation (temazepam formation), using liver microsomes obtained from extensive (EM) and poor metabolizers (PM) for S-mephenytoin 4"-hydroxylation. Nordazepam 124-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 7704038-1 1994 Metabolism of diazepam was studied in vitro to identify the forms of cytochrome P450 (CYP) responsible for N-demethylation (nordazepam formation) and 3-hydroxylation (temazepam formation), using liver microsomes obtained from extensive (EM) and poor metabolizers (PM) for S-mephenytoin 4"-hydroxylation. Temazepam 167-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 7704038-1 1994 Metabolism of diazepam was studied in vitro to identify the forms of cytochrome P450 (CYP) responsible for N-demethylation (nordazepam formation) and 3-hydroxylation (temazepam formation), using liver microsomes obtained from extensive (EM) and poor metabolizers (PM) for S-mephenytoin 4"-hydroxylation. Temazepam 167-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 7704038-1 1994 Metabolism of diazepam was studied in vitro to identify the forms of cytochrome P450 (CYP) responsible for N-demethylation (nordazepam formation) and 3-hydroxylation (temazepam formation), using liver microsomes obtained from extensive (EM) and poor metabolizers (PM) for S-mephenytoin 4"-hydroxylation. Mephenytoin 272-285 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-84 7704038-1 1994 Metabolism of diazepam was studied in vitro to identify the forms of cytochrome P450 (CYP) responsible for N-demethylation (nordazepam formation) and 3-hydroxylation (temazepam formation), using liver microsomes obtained from extensive (EM) and poor metabolizers (PM) for S-mephenytoin 4"-hydroxylation. Mephenytoin 272-285 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-89 7895609-0 1994 Cytochrome P450 isozymes involved in propranolol metabolism in human liver microsomes. Propranolol 37-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 7895609-3 1994 We characterized cytochrome P450 isozymes responsible for propranolol metabolism, especially N-desisopropylation and 5-hydroxylation, in human liver microsomes. Propranolol 58-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-32 7895609-3 1994 We characterized cytochrome P450 isozymes responsible for propranolol metabolism, especially N-desisopropylation and 5-hydroxylation, in human liver microsomes. Nitrogen 93-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-32 7959460-3 1994 A group of 19 acyclic terpenes have been evaluated for potential toxicity/carcinogenicity by molecular orbital determinations of their spatial and electronic parameters, and hence prediction of their metabolic activation or detoxication by the cytochrome P-450 (CYP) superfamily of mixed-function oxidase enzymes. Terpenes 22-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 244-260 7959460-3 1994 A group of 19 acyclic terpenes have been evaluated for potential toxicity/carcinogenicity by molecular orbital determinations of their spatial and electronic parameters, and hence prediction of their metabolic activation or detoxication by the cytochrome P-450 (CYP) superfamily of mixed-function oxidase enzymes. Terpenes 22-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-265 7532811-7 1994 As a cytochrome P-450, NOS catalyzes a heme-mediated reduction of molecular oxygen, resulting in the formation of H2O2 in the absence of L-arginine. Heme 39-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 7532811-7 1994 As a cytochrome P-450, NOS catalyzes a heme-mediated reduction of molecular oxygen, resulting in the formation of H2O2 in the absence of L-arginine. Oxygen 76-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 7532811-7 1994 As a cytochrome P-450, NOS catalyzes a heme-mediated reduction of molecular oxygen, resulting in the formation of H2O2 in the absence of L-arginine. Hydrogen Peroxide 114-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 7532811-7 1994 As a cytochrome P-450, NOS catalyzes a heme-mediated reduction of molecular oxygen, resulting in the formation of H2O2 in the absence of L-arginine. Arginine 137-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 7695156-9 1995 These results indicate that cytochrome P-450 metabolic pathways have an important role in propofol clearance and propofol anesthetic recovery in Greyhounds. Propofol 90-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 7695156-9 1995 These results indicate that cytochrome P-450 metabolic pathways have an important role in propofol clearance and propofol anesthetic recovery in Greyhounds. Propofol 113-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 8846617-10 1995 The specific cytochrome P450 (CYP) isoenzymes involved in the metabolism of fluvoxamine are unknown. Fluvoxamine 76-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 8846617-10 1995 The specific cytochrome P450 (CYP) isoenzymes involved in the metabolism of fluvoxamine are unknown. Fluvoxamine 76-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-33 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Venlafaxine Hydrochloride 56-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Venlafaxine Hydrochloride 56-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Sparteine 204-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Sparteine 204-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Dextromethorphan 221-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Dextromethorphan 221-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Paroxetine 250-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Paroxetine 250-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Fluoxetine 266-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Fluoxetine 266-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. norfluoxetine 290-303 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. norfluoxetine 290-303 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Sertraline 314-324 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Sertraline 314-324 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-153 8846618-2 1995 The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine identical to norfluoxetine > or = sertraline > or = fluvoxamine > venlafaxine. Fluvoxamine 335-346 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 7713363-11 1995 DL decreased the PB-induced BND activity and cytochrome P-450 and cytochrome b5 content. Phenobarbital 17-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-79 7745225-3 1995 CCl4-induced changes in hepatic biochemical parameters by reducing cytochrome P-450, by comparing the effects of colchicine and SKF 525-A, a well-known inhibitor of cytochrome P-450. Proadifen 128-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-181 7532106-3 1994 These studies involved the use of the imidazole antimycotics, econazole and miconazole, which are inhibitors of cytochrome P-450. imidazole 38-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 7532106-3 1994 These studies involved the use of the imidazole antimycotics, econazole and miconazole, which are inhibitors of cytochrome P-450. Econazole 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 7532106-3 1994 These studies involved the use of the imidazole antimycotics, econazole and miconazole, which are inhibitors of cytochrome P-450. Miconazole 76-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 8082935-1 1994 The catabolism of various calcium channel blockers through cytochrome P-450 is heterogeneous and may be modified by concomitant use of cyclosporin A. Cyclosporine 135-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 7873467-6 1994 Both the relaxing effect on vascular smooth muscle and the effect on platelets are considered to be due to a stimulation of soluble guanylate cyclase by nitric oxide derived from the organic nitrate ester molecule through metabolization catalyzed by enzymes such as glutathione S-transferase, cytochrome P-450, and possibly esterases. Nitric Oxide 153-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 293-309 7873467-6 1994 Both the relaxing effect on vascular smooth muscle and the effect on platelets are considered to be due to a stimulation of soluble guanylate cyclase by nitric oxide derived from the organic nitrate ester molecule through metabolization catalyzed by enzymes such as glutathione S-transferase, cytochrome P-450, and possibly esterases. nitrate ester 191-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 293-309 7949373-1 1994 Metabolic N-dealkylation is a commonly observed biotransformation with tertiary and secondary amine drugs and related N-alkylated amides, but surprisingly little is known about the cytochrome P-450 isozymes involved in these dealkylation reactions. Nitrogen 10-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 8052309-0 1994 An efficient mimic of cytochrome P-450 from a zeolite-encaged iron complex in a polymer membrane. Zeolites 46-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 8052309-0 1994 An efficient mimic of cytochrome P-450 from a zeolite-encaged iron complex in a polymer membrane. Iron 62-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 8052309-0 1994 An efficient mimic of cytochrome P-450 from a zeolite-encaged iron complex in a polymer membrane. Polymers 80-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 8052309-7 1994 The polymer acts as a mimic of the phospholipid membrane in which cytochrome P-450 resides, acting as an interface between two immiscible phases and avoiding the need for solvents or phase-transfer agents. Polymers 4-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 8052309-7 1994 The polymer acts as a mimic of the phospholipid membrane in which cytochrome P-450 resides, acting as an interface between two immiscible phases and avoiding the need for solvents or phase-transfer agents. Phospholipids 35-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 8068004-6 1994 Consistent with this idea, the imidazole cytochrome P-450 inhibitors miconazole, econazole, clotrimazole and ketoconazole inhibited the thapsigargin-elevated [Ca2+]i with pIC50 values of 7.1, 7.1, 7.1 and 5.8 respectively. Miconazole 69-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 8068004-6 1994 Consistent with this idea, the imidazole cytochrome P-450 inhibitors miconazole, econazole, clotrimazole and ketoconazole inhibited the thapsigargin-elevated [Ca2+]i with pIC50 values of 7.1, 7.1, 7.1 and 5.8 respectively. Econazole 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 8068004-6 1994 Consistent with this idea, the imidazole cytochrome P-450 inhibitors miconazole, econazole, clotrimazole and ketoconazole inhibited the thapsigargin-elevated [Ca2+]i with pIC50 values of 7.1, 7.1, 7.1 and 5.8 respectively. Clotrimazole 92-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 8068004-6 1994 Consistent with this idea, the imidazole cytochrome P-450 inhibitors miconazole, econazole, clotrimazole and ketoconazole inhibited the thapsigargin-elevated [Ca2+]i with pIC50 values of 7.1, 7.1, 7.1 and 5.8 respectively. Ketoconazole 109-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 8068004-6 1994 Consistent with this idea, the imidazole cytochrome P-450 inhibitors miconazole, econazole, clotrimazole and ketoconazole inhibited the thapsigargin-elevated [Ca2+]i with pIC50 values of 7.1, 7.1, 7.1 and 5.8 respectively. Thapsigargin 136-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 8068004-9 1994 We examined whether the inhibition of Ca2+ influx was due to an interaction of the inhibitor imidazole nitrogen with the haem iron of the putative cytochrome P-450 by comparing the activity of two compounds, identical except that one was methylated at the imidazole 2-position. imidazole nitrogen 93-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 8068004-9 1994 We examined whether the inhibition of Ca2+ influx was due to an interaction of the inhibitor imidazole nitrogen with the haem iron of the putative cytochrome P-450 by comparing the activity of two compounds, identical except that one was methylated at the imidazole 2-position. Iron 126-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 8068004-9 1994 We examined whether the inhibition of Ca2+ influx was due to an interaction of the inhibitor imidazole nitrogen with the haem iron of the putative cytochrome P-450 by comparing the activity of two compounds, identical except that one was methylated at the imidazole 2-position. imidazole 93-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 7986010-7 1994 A possible explanation of drug resistance based on the observation of suicide inactivation of bacterial cytochrome P-450 by the cyclopropylamine moiety has also been proposed and is discussed in this report. cyclopropylamine 128-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 7981013-2 1994 The primary metabolism of diazepam was studied in human liver microsomes in order to investigate the kinetics and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of the main diazepam metabolites, temazepam and N-desmethyldiazepam. Diazepam 26-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-145 7981013-2 1994 The primary metabolism of diazepam was studied in human liver microsomes in order to investigate the kinetics and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of the main diazepam metabolites, temazepam and N-desmethyldiazepam. Diazepam 26-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 7981013-2 1994 The primary metabolism of diazepam was studied in human liver microsomes in order to investigate the kinetics and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of the main diazepam metabolites, temazepam and N-desmethyldiazepam. Diazepam 203-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-145 7981013-2 1994 The primary metabolism of diazepam was studied in human liver microsomes in order to investigate the kinetics and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of the main diazepam metabolites, temazepam and N-desmethyldiazepam. Diazepam 203-211 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 7981013-2 1994 The primary metabolism of diazepam was studied in human liver microsomes in order to investigate the kinetics and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of the main diazepam metabolites, temazepam and N-desmethyldiazepam. Temazepam 225-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-145 7981013-2 1994 The primary metabolism of diazepam was studied in human liver microsomes in order to investigate the kinetics and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of the main diazepam metabolites, temazepam and N-desmethyldiazepam. Temazepam 225-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 7981013-2 1994 The primary metabolism of diazepam was studied in human liver microsomes in order to investigate the kinetics and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of the main diazepam metabolites, temazepam and N-desmethyldiazepam. Nordazepam 239-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-145 7981013-2 1994 The primary metabolism of diazepam was studied in human liver microsomes in order to investigate the kinetics and to identify the cytochrome P450 (CYP) isoforms responsible for the formation of the main diazepam metabolites, temazepam and N-desmethyldiazepam. Nordazepam 239-258 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 7981013-11 1994 Studies with a series of CYP isoform selective inhibitors and with an inhibitory anti-CYP2C antibody indicated that temazepam formation was carried out mainly by CYP3A isoforms, whereas the formation of N-desmethyldiazepam was mediated by both CYP3A isoforms and S-mephenytoin hydroxylase. Temazepam 116-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-28 7935340-1 1994 Bufuralol 1"-hydroxylation is a prototypical reaction catalyzed by cytochrome P450 (P450) 2D6, an enzyme known to show debrisoquine/sparteine-type genetic polymorphism in humans. bufuralol 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-93 7935340-1 1994 Bufuralol 1"-hydroxylation is a prototypical reaction catalyzed by cytochrome P450 (P450) 2D6, an enzyme known to show debrisoquine/sparteine-type genetic polymorphism in humans. Debrisoquin 119-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-93 8037457-1 1994 Human cytochrome P450 (P450) 1A1 is primarily an extrahepatic enzyme and is important because of its roles in the activation of polycyclic hydrocarbons and other xenobiotic chemicals. Hydrocarbons, Cyclic 128-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-32 7917780-0 1994 Identification of human liver cytochrome P450 isoforms mediating secondary omeprazole metabolism. Omeprazole 75-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 7519397-1 1994 Ketoconazole, cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate, and gossypol are reported inhibitors of the lipoxygenase (LO) and cytochrome P-450 enzyme systems and are potent blockers of swelling-activated efflux of organic osmolytes and volume-sensitive anion channels in C6 glioma cells. Ketoconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 7519397-1 1994 Ketoconazole, cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate, and gossypol are reported inhibitors of the lipoxygenase (LO) and cytochrome P-450 enzyme systems and are potent blockers of swelling-activated efflux of organic osmolytes and volume-sensitive anion channels in C6 glioma cells. CINNAMYL-3,4-DIHYDROXY-ALPHA-CYANOCINNAMATE 14-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 7519397-1 1994 Ketoconazole, cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate, and gossypol are reported inhibitors of the lipoxygenase (LO) and cytochrome P-450 enzyme systems and are potent blockers of swelling-activated efflux of organic osmolytes and volume-sensitive anion channels in C6 glioma cells. Gossypol 63-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 7519397-2 1994 To directly test the hypothesis that LO- or cytochrome P-450-derived products of arachidonic acid (AA) participate in the regulation of these volume-sensitive transport pathways, we incubated C6 cells with [1-14C]AA and observed the extent and profile of its conversion under basal conditions and after acute swelling. Arachidonic Acid 81-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-60 7948817-3 1994 Cytochrome P450 content was reduced to 17% of control values after acute carbon tetrachloride and to 35% of control values after bile duct ligation. Carbon Tetrachloride 73-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. Troleandomycin 34-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-205 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. Troleandomycin 34-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-210 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. Troleandomycin 57-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-205 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. Troleandomycin 57-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-210 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. alpha-naphthoflavone 63-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-205 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. alpha-naphthoflavone 63-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-210 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. Ditiocarb 95-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-205 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. Ditiocarb 95-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-210 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. Ditiocarb 119-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 190-205 8203907-1 1994 A variety of chemicals, including triacetyloleandomycin (TAO), alpha-naphthoflavone (ANF), and diethyldithiocarbamate (DDC), are widely used as inhibitory probes for select individual human cytochrome P450 (CYP) enzymes, despite the fact that the selectivity of these inhibitors has not been rigorously evaluated. Ditiocarb 119-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 207-210 7917780-2 1994 The in vitro metabolism of omeprazole was studied in human liver microsomes in order to define the secondary metabolic pathways and identify the cytochrome P450 (CYP) isoforms responsible for the formation of the secondary metabolites of omeprazole. Omeprazole 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 7917780-2 1994 The in vitro metabolism of omeprazole was studied in human liver microsomes in order to define the secondary metabolic pathways and identify the cytochrome P450 (CYP) isoforms responsible for the formation of the secondary metabolites of omeprazole. Omeprazole 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 7917780-2 1994 The in vitro metabolism of omeprazole was studied in human liver microsomes in order to define the secondary metabolic pathways and identify the cytochrome P450 (CYP) isoforms responsible for the formation of the secondary metabolites of omeprazole. Omeprazole 238-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-160 8081318-5 1994 The demethylation of caffeine by the hepatic cytochrome P-450 oxygenases begins within minutes and dimethylxanthines (especially paraxanthine) are generated. Caffeine 21-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 7917780-2 1994 The in vitro metabolism of omeprazole was studied in human liver microsomes in order to define the secondary metabolic pathways and identify the cytochrome P450 (CYP) isoforms responsible for the formation of the secondary metabolites of omeprazole. Omeprazole 238-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-165 8081318-5 1994 The demethylation of caffeine by the hepatic cytochrome P-450 oxygenases begins within minutes and dimethylxanthines (especially paraxanthine) are generated. Theophylline 99-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 8081318-5 1994 The demethylation of caffeine by the hepatic cytochrome P-450 oxygenases begins within minutes and dimethylxanthines (especially paraxanthine) are generated. 1,7-dimethylxanthine 129-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 7917780-6 1994 The formation kinetics of these two metabolites from omeprazole sulphone were biphasic suggesting the involvement of multiple CYP isoforms in each case. omeprazole sulfone 53-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-129 8043490-15 1994 Non-steroidal inhibitors of cytochrome P-450 enzymes, such as imidazole and triazole derivatives have been developed which are highly selective for aromatase. imidazole 62-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 8043490-15 1994 Non-steroidal inhibitors of cytochrome P-450 enzymes, such as imidazole and triazole derivatives have been developed which are highly selective for aromatase. Triazoles 76-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 8022422-1 1994 The antihypertensive drug dihydralazine may, on rare occasions, cause immunoallergic hepatitis characterized by anti-cytochrome P450 (P450)1A2 autoantibodies. Dihydralazine 26-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-142 8054246-2 1994 The activation of proguanil to cycloguanil by human liver microsomes was studied to define the cytochrome P450 (CYP) isoforms involved in this reaction. Proguanil 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-110 8175769-0 1994 NADPH-initiated cytochrome P450-mediated free metal ion-independent oxidative damage of microsomal proteins. NADP 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 8175769-0 1994 NADPH-initiated cytochrome P450-mediated free metal ion-independent oxidative damage of microsomal proteins. Metals 46-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 8175769-2 1994 In this paper we demonstrate that NADPH-initiated oxidative damage of microsomal proteins occurs in the absence of free metal ions and that this protein oxidation is mediated by cytochrome P450 (cyt P450). NADP 34-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 178-193 8175769-2 1994 In this paper we demonstrate that NADPH-initiated oxidative damage of microsomal proteins occurs in the absence of free metal ions and that this protein oxidation is mediated by cytochrome P450 (cyt P450). NADP 34-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-203 8175769-4 1994 Ascorbate (AH2) specifically inhibits free metal ion-independent cyt P450-mediated protein oxidation and thereby prevents subsequent proteolytic degradation. Ascorbic Acid 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-73 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. ammonium ferrous sulfate 65-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-8 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. ammonium ferrous sulfate 65-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-42 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. ammonium ferrous sulfate 65-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. ammonium ferrous sulfate 65-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. ammonium ferrous sulfate 65-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 86-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-8 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 86-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-42 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 86-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 86-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 86-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. fe2+ o2 144-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-8 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. fe2+ o2 144-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-42 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. fe2+ o2 144-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. fe2+ o2 144-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. fe2+ o2 144-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. perferryl 186-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-8 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. perferryl 186-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-42 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. perferryl 186-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. perferryl 186-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. perferryl 186-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 149-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-8 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 149-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-42 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 149-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 149-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 8175769-9 1994 Cyt P450 Fe3+ is reduced by NADPH-cyt P450 reductase to cyt P450 Fe2+, which consumes oxygen in a stoichiometric proportion to produce cyt P450 Fe2+ O2, the resonance form of which is a perferryl moiety, cyt P450 Fe3+.O2-.. Oxygen 149-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-64 7938083-0 1994 5-Aminosalicylic acid inhibits leukotriene B4 omega-hydroxylase activity in human polymorphonuclear leukocytes. Mesalamine 0-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-63 7938083-2 1994 To investigate the action of 5-aminosalicylic acid (5-ASA) on LTB4 omega-hydroxylase activity, we incubated human polymorphonuclear leukocytes (PMNLs) with 3H-labeled LTB4 after pre-incubation with various concentrations of 5-ASA. Mesalamine 52-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-84 7938083-4 1994 LTB4 omega-hydroxylase activity was inhibited by 5-ASA in a concentration-dependent fashion. Mesalamine 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-22 8054246-2 1994 The activation of proguanil to cycloguanil by human liver microsomes was studied to define the cytochrome P450 (CYP) isoforms involved in this reaction. Proguanil 18-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-115 8054246-2 1994 The activation of proguanil to cycloguanil by human liver microsomes was studied to define the cytochrome P450 (CYP) isoforms involved in this reaction. cycloguanil 31-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-115 8207335-8 1994 Corneal epithelial hypoxia in response to contact lens wear results in the time-dependent formation of NADPH-cytochrome P450-dependent arachidonate metabolites, 12(R)-HETE and 12(R)-HETrE. (r)-hetre 178-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 8205227-11 1994 Eugenol elicited type 1 changes in the spectrum of microsomal cytochrome P-450. Eugenol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 8205227-13 1994 Eugenol significantly protected against the degradation of cytochrome P-450 during lipid peroxidation with all the systems tested. Eugenol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 7926597-0 1994 Modification of testicular cytochrome P-450 after fenitrothion administration. Fenitrothion 50-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 7926597-4 1994 Acute (165 mg/kg, 1 day) and subacute (55 mg/kg, 3 days) administration of fenitrothion caused a significant decrease on testicular cytochrome P-450 content (51 and 50% respectively) but no modification on cytochrome b5 and NADPH cytochrome c reductase. Fenitrothion 75-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 8207335-1 1994 The corneal epithelium of several species, has the capacity to metabolize arachidonic acid (arachidonic acid) via an NADPH-dependent cytochrome P450 mechanism. Arachidonic Acid 74-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8207335-1 1994 The corneal epithelium of several species, has the capacity to metabolize arachidonic acid (arachidonic acid) via an NADPH-dependent cytochrome P450 mechanism. Arachidonic Acid 92-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 8207335-6 1994 The formation of these metabolites is unaffected by cyclooxygenase and lipoxygenase inhibitors (indomethacin, diclofenac and BW755C) but inhibited by cytochrome P450 enzyme inhibitors such as carbon monoxide, SKF-525A and clotrimazole. Carbon Monoxide 192-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-165 8207335-10 1994 It is evident that the cytochrome P450 arachidonate metabolites should be added to the realm of cyclooxygenase and lipoxygenase-derived eicosanoids as possible inflammatory mediators. Arachidonic Acid 39-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-38 8207335-6 1994 The formation of these metabolites is unaffected by cyclooxygenase and lipoxygenase inhibitors (indomethacin, diclofenac and BW755C) but inhibited by cytochrome P450 enzyme inhibitors such as carbon monoxide, SKF-525A and clotrimazole. Proadifen 209-217 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-165 8173131-3 1994 DATA SYNTHESIS: Fluconazole is a broad-spectrum triazole antifungal agent primarily eliminated by renal mechanisms, although hepatic cytochrome P-450 inhibition and hepatotoxicity have been observed. Fluconazole 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-149 8207335-6 1994 The formation of these metabolites is unaffected by cyclooxygenase and lipoxygenase inhibitors (indomethacin, diclofenac and BW755C) but inhibited by cytochrome P450 enzyme inhibitors such as carbon monoxide, SKF-525A and clotrimazole. Clotrimazole 222-234 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-165 8207335-7 1994 The capacity of the normal corneal epithelium to metabolize arachidonic acid via cytochrome P450 is very low although under certain conditions this enzymatic pathway may become greatly induced. Arachidonic Acid 60-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 8207335-8 1994 Corneal epithelial hypoxia in response to contact lens wear results in the time-dependent formation of NADPH-cytochrome P450-dependent arachidonate metabolites, 12(R)-HETE and 12(R)-HETrE. NADP 103-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 8207335-8 1994 Corneal epithelial hypoxia in response to contact lens wear results in the time-dependent formation of NADPH-cytochrome P450-dependent arachidonate metabolites, 12(R)-HETE and 12(R)-HETrE. Arachidonic Acid 135-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 8207335-8 1994 Corneal epithelial hypoxia in response to contact lens wear results in the time-dependent formation of NADPH-cytochrome P450-dependent arachidonate metabolites, 12(R)-HETE and 12(R)-HETrE. 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid 161-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-124 7907372-6 1994 Thus the cytochrome P-450-mediated biotransformation of taxol to 6 alpha-hydroxytaxol can be classified as a detoxification reaction. Paclitaxel 56-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 7907372-6 1994 Thus the cytochrome P-450-mediated biotransformation of taxol to 6 alpha-hydroxytaxol can be classified as a detoxification reaction. alpha-hydroxytaxol 67-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 8160800-6 1994 Indomethacin, 10 microM, an inhibitor of AA-dependent cyclooxygenase, did not prevent the PTH action, but 2 microM 7-ethoxyresorufin, a cytochrome P-450 inhibitor, prevented the PTH effect. ethoxyresorufin 115-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-152 8158054-2 1994 In an attempt to inhibit cytochrome P-450 metabolism of praziquantel, cimetidine (1600 mg/day) was coadministered. Praziquantel 56-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 8171446-5 1994 The irreversible binding of both these DDT metabolites was inhibited by metyrapone, indicating the involvement of cytochrome P-450. DDT 39-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 8171446-5 1994 The irreversible binding of both these DDT metabolites was inhibited by metyrapone, indicating the involvement of cytochrome P-450. Metyrapone 72-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 8049635-2 1994 Debrisoquine metabolism is controlled by a cytochrome P-450 isozyme encoded at the CYP2D6 locus, which is inducible by antipyrine and rifampicin. Debrisoquin 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 8049635-2 1994 Debrisoquine metabolism is controlled by a cytochrome P-450 isozyme encoded at the CYP2D6 locus, which is inducible by antipyrine and rifampicin. Antipyrine 119-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 8049635-2 1994 Debrisoquine metabolism is controlled by a cytochrome P-450 isozyme encoded at the CYP2D6 locus, which is inducible by antipyrine and rifampicin. Rifampin 134-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 8013290-0 1994 Cytochrome P-450-dependent biotransformation of 2-acetylaminofluorene in cell-free preparations of human embryonic hepatic, adrenal, renal, pulmonary, and cardiac tissues. 2-Acetylaminofluorene 48-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8304420-6 1994 Together with previous results, these observations indicate that 1) epidermis generates 12-HETE by either cytochrome P-450 or lipoxygenase-based mechanisms depending on reaction conditions, and 2) 12-lipoxygenases (originally described in hematopoietic cell types) may be expressed in at least two distinct isoforms in epithelial barriers in humans, and in the case of the skin, a microsomal (platelet-type) 12-lipoxygenase is selectively overexpressed in germinal layer keratinocytes during psoriatic inflammation. 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid 88-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-138 8286397-2 1994 In contrast, the electronic absorption spectra for camphor-free cytochrome P-450cam between 78 K and 295 K show no significant spectral changes. Camphor 51-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 7903909-0 1994 Taxol metabolism by human liver microsomes: identification of cytochrome P450 isozymes involved in its biotransformation. Paclitaxel 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-77 8171214-5 1994 The enzyme system responsible for PAC activation is the mixed-function oxidase system and, in particular, cytochrome P-450. pac 34-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 8082573-2 1994 Inherent substrate factors, such as basicity, electronic state, lipophilicity, and conformation control binding of the diverse classes of amines to cytochrome P-450. Amines 138-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 8082573-6 1994 Certain amines appear to regulate O2 association with cytochrome P-450 and stabilize the various oxy species formed. Amines 8-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 8082573-6 1994 Certain amines appear to regulate O2 association with cytochrome P-450 and stabilize the various oxy species formed. Oxygen 34-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 8082573-7 1994 Considering the selective prerequisites for oxidative attack by cytochrome P-450 at vulnerable nitrogen centers, many cytotoxic amines belonging to the category of relatively rigid, planar molecules undergo N-oxidative activation by the cytochrome P-450IA subfamily, while more bulky amines with flexible conformation are N-oxygenated preferentially by phenobarbital-inducible cytochromes P-450. Nitrogen 95-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 8082573-7 1994 Considering the selective prerequisites for oxidative attack by cytochrome P-450 at vulnerable nitrogen centers, many cytotoxic amines belonging to the category of relatively rigid, planar molecules undergo N-oxidative activation by the cytochrome P-450IA subfamily, while more bulky amines with flexible conformation are N-oxygenated preferentially by phenobarbital-inducible cytochromes P-450. Amines 128-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 8082573-7 1994 Considering the selective prerequisites for oxidative attack by cytochrome P-450 at vulnerable nitrogen centers, many cytotoxic amines belonging to the category of relatively rigid, planar molecules undergo N-oxidative activation by the cytochrome P-450IA subfamily, while more bulky amines with flexible conformation are N-oxygenated preferentially by phenobarbital-inducible cytochromes P-450. Nitrogen 207-208 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 8082573-7 1994 Considering the selective prerequisites for oxidative attack by cytochrome P-450 at vulnerable nitrogen centers, many cytotoxic amines belonging to the category of relatively rigid, planar molecules undergo N-oxidative activation by the cytochrome P-450IA subfamily, while more bulky amines with flexible conformation are N-oxygenated preferentially by phenobarbital-inducible cytochromes P-450. Amines 284-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 8082573-7 1994 Considering the selective prerequisites for oxidative attack by cytochrome P-450 at vulnerable nitrogen centers, many cytotoxic amines belonging to the category of relatively rigid, planar molecules undergo N-oxidative activation by the cytochrome P-450IA subfamily, while more bulky amines with flexible conformation are N-oxygenated preferentially by phenobarbital-inducible cytochromes P-450. Phenobarbital 353-366 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 8082573-8 1994 Small differences in protein structure between the various cytochrome P-450 subforms might serve to stabilize aminium radicals to permit oxygen rebound. aminium 110-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 8082573-8 1994 Small differences in protein structure between the various cytochrome P-450 subforms might serve to stabilize aminium radicals to permit oxygen rebound. Oxygen 137-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 7957514-6 1994 The results indicate that fluconazole is a minor inhibitor of theophylline disposition compared with enoxacin, and they suggest that the inhibitory action of fluconazole is selective for certain cytochrome P-450 isozymes, but not for the cytochrome P-4501A involved in theophylline metabolism. Fluconazole 158-169 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 195-211 8123970-5 1994 DATA SYNTHESIS: Ciprofloxacin has been reported to interact with cyclosporine during concomitant use through an interaction with the cytochrome P-450 system or by additive nephrotoxicity. Ciprofloxacin 16-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-149 8123970-5 1994 DATA SYNTHESIS: Ciprofloxacin has been reported to interact with cyclosporine during concomitant use through an interaction with the cytochrome P-450 system or by additive nephrotoxicity. Cyclosporine 65-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-149 7544830-0 1994 Induction of cytochrome P-450 by 5-methoxypsoralen in the yeast Saccharomyces cerevisiae. 5-Methoxypsoralen 33-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 7544830-1 1994 Yeast cells (D7 strain) incubated in the presence of 5-methoxypsoralen (5-MOP) increase the activity of the monooxygenase system cytochrome P-450 dependent (cytochrome P-450 level and 7-ethoxycoumarin-O-diethylase activity). 5-Methoxypsoralen 53-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 7544830-1 1994 Yeast cells (D7 strain) incubated in the presence of 5-methoxypsoralen (5-MOP) increase the activity of the monooxygenase system cytochrome P-450 dependent (cytochrome P-450 level and 7-ethoxycoumarin-O-diethylase activity). 5-Methoxypsoralen 53-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 7544830-1 1994 Yeast cells (D7 strain) incubated in the presence of 5-methoxypsoralen (5-MOP) increase the activity of the monooxygenase system cytochrome P-450 dependent (cytochrome P-450 level and 7-ethoxycoumarin-O-diethylase activity). 5-Methoxypsoralen 72-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-145 7544830-1 1994 Yeast cells (D7 strain) incubated in the presence of 5-methoxypsoralen (5-MOP) increase the activity of the monooxygenase system cytochrome P-450 dependent (cytochrome P-450 level and 7-ethoxycoumarin-O-diethylase activity). 5-Methoxypsoralen 72-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 7544830-4 1994 The capacity of 5-MOP to induce the cytochrome P-450 system in eukaryotic cells, in which it is known to be involved in the metabolism of the psoralen, may decrease the availability of the compound for photo-induced genotoxic reactions, which may explain the good tolerance in patients. 5-Methoxypsoralen 16-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-52 7808019-8 1994 Both of these hypercatabolic states can be modulated by concurrent administration of ketoconazole, an inhibitor of cytochrome P-450 and lipoxygenase-mediated oxidations. Ketoconazole 85-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 115-148 7504205-13 1993 The nutritional effect of vitamin E on hepatic microsomal cytochrome P-450 activities modified the AFB1 genotoxicity measured in vitro. Vitamin E 26-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 8242617-1 1993 The present study identifies the specific human cytochrome P-450 (CYP) enzymes involved in hydroxylation leading to activation of the anticancer drug cyclophosphamide and its isomeric analogue, ifosphamide. Cyclophosphamide 150-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 8242617-1 1993 The present study identifies the specific human cytochrome P-450 (CYP) enzymes involved in hydroxylation leading to activation of the anticancer drug cyclophosphamide and its isomeric analogue, ifosphamide. Cyclophosphamide 150-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 8242617-1 1993 The present study identifies the specific human cytochrome P-450 (CYP) enzymes involved in hydroxylation leading to activation of the anticancer drug cyclophosphamide and its isomeric analogue, ifosphamide. Ifosfamide 194-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 8242617-1 1993 The present study identifies the specific human cytochrome P-450 (CYP) enzymes involved in hydroxylation leading to activation of the anticancer drug cyclophosphamide and its isomeric analogue, ifosphamide. Ifosfamide 194-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-69 8242617-8 1993 Furthermore, growth of cultured CYP2A6- and CYP2B6-expressing B-lymphoblastoid cells, but not of CYP-negative control cells, was inhibited by cyclophosphamide and ifosphamide as a consequence of prodrug activation to cytotoxic metabolites. Cyclophosphamide 142-158 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 8242617-8 1993 Furthermore, growth of cultured CYP2A6- and CYP2B6-expressing B-lymphoblastoid cells, but not of CYP-negative control cells, was inhibited by cyclophosphamide and ifosphamide as a consequence of prodrug activation to cytotoxic metabolites. Ifosfamide 163-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 8307581-1 1993 Human CYP2E encodes an ethanol-inducible cytochrome P450 monooxygenase that metabolizes various carcinogens and may therefore play a role in cancer susceptibility. Ethanol 23-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-56 8230282-3 1993 We speculated that the decline in drug levels, indicating acquired resistance, resulted partly from inducible cytochrome-P450 oxidative enzymes, which can catabolize all-trans RA. Radium 176-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-125 8230282-4 1993 PURPOSE: We studied the clinical pharmacology of all-trans RA in cancer patients to determine possible mechanisms of acquired resistance and evaluated the potential for reversal by ketoconazole, an inhibitor of cytochrome-P450 oxidative enzymes. Ketoconazole 181-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-226 8230282-14 1993 Ketoconazole attenuates this accelerated catabolism, suggesting that oxidation by cytochrome-P450 enzymes is an important pathway for both constitutive and induced pathways of all-trans RA metabolism. Ketoconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 8230282-14 1993 Ketoconazole attenuates this accelerated catabolism, suggesting that oxidation by cytochrome-P450 enzymes is an important pathway for both constitutive and induced pathways of all-trans RA metabolism. Radium 186-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 12959268-0 1993 Identification of human liver cytochrome P450 isoforms mediating omeprazole metabolism. Omeprazole 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 12959268-1 1993 1 The in vitro metabolism of omeprazole was studied in human liver microsomes in order to define the metabolic pathways and identify the cytochrome P450 (CYP) isoforms responsible for the formation of the major omeprazole metabolites. Omeprazole 29-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-157 12959268-1 1993 1 The in vitro metabolism of omeprazole was studied in human liver microsomes in order to define the metabolic pathways and identify the cytochrome P450 (CYP) isoforms responsible for the formation of the major omeprazole metabolites. Omeprazole 211-221 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 154-157 7504205-13 1993 The nutritional effect of vitamin E on hepatic microsomal cytochrome P-450 activities modified the AFB1 genotoxicity measured in vitro. Aflatoxin B1 99-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 8240292-0 1993 Stimulation by paraquat of microsomal and cytochrome P-450-dependent oxidation of glycerol to formaldehyde. Glycerol 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 8240292-0 1993 Stimulation by paraquat of microsomal and cytochrome P-450-dependent oxidation of glycerol to formaldehyde. Formaldehyde 94-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 8240292-1 1993 Glycerol can be oxidized to formaldehyde by microsomes in a reaction that is dependent on cytochrome P-450. Glycerol 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 8240292-1 1993 Glycerol can be oxidized to formaldehyde by microsomes in a reaction that is dependent on cytochrome P-450. Formaldehyde 28-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 8240292-11 1993 However, cytochrome P-450 is required for elevated rates of formaldehyde production even in the presence of paraquat. Formaldehyde 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 8240292-11 1993 However, cytochrome P-450 is required for elevated rates of formaldehyde production even in the presence of paraquat. Paraquat 108-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 7905377-5 1993 Biotransformation of lovastatin occurred by three distinct routes, namely hydrolysis of the lactone ring to yield the pharmacologically active dihydroxy acid, cytochrome P-450-mediated oxidation of the fused-ring system, and beta-oxidation of the dihydroxy acid side chain. Lovastatin 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 159-175 7905377-11 1993 From these studies, cytochrome P-450 oxidation is the primary route of phase I metabolism for lovastatin in human and dog, but beta-oxidation plays a major metabolic role in rodents. Lovastatin 94-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 8143890-1 1993 Estrogens are synthesized from C19 steroids by a unique form of cytochrome P450, aromatase cytochrome P-450 (P-450AROM; the product of the CYP19 gene). Steroids 35-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-107 8111292-6 1993 This indicates that the metabolism of aflatoxin B1 brought about by the cytochrome P-450-dependent monooxygenase system plays an important role in the action of aflatoxin B1 on human lymphocytes. Aflatoxin B1 38-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 8111292-6 1993 This indicates that the metabolism of aflatoxin B1 brought about by the cytochrome P-450-dependent monooxygenase system plays an important role in the action of aflatoxin B1 on human lymphocytes. Aflatoxin B1 161-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 8310712-2 1993 A commercially-available human cytochrome P450 isozyme (CYP2D6) preparation was used in imipramine metabolism studies. Imipramine 88-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 8240252-1 1993 Cytochrome P-450 has been suggested as a mediator of the signal between depleted platelet calcium stores and an increase in plasma membrane permeability to calcium which follows depletion of the stores. Calcium 90-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8240252-1 1993 Cytochrome P-450 has been suggested as a mediator of the signal between depleted platelet calcium stores and an increase in plasma membrane permeability to calcium which follows depletion of the stores. Calcium 156-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8240252-8 1993 Inhibition of cytochrome P-450 with CO bubbled through platelet suspensions did not change calcium influx in resting cells and potentiated TG-induced calcium influx (160% of control calcium accumulation at 20 min). Thapsigargin 139-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 8240252-8 1993 Inhibition of cytochrome P-450 with CO bubbled through platelet suspensions did not change calcium influx in resting cells and potentiated TG-induced calcium influx (160% of control calcium accumulation at 20 min). Calcium 150-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 8240252-2 1993 This hypothesis is based on the observations that inhibitors of cytochrome P-450, such as the imidazole antifungal agents, also inhibit influx of a calcium surrogate (manganese) into calcium-depleted platelets. imidazole 94-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 8240252-8 1993 Inhibition of cytochrome P-450 with CO bubbled through platelet suspensions did not change calcium influx in resting cells and potentiated TG-induced calcium influx (160% of control calcium accumulation at 20 min). Calcium 150-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 8240252-2 1993 This hypothesis is based on the observations that inhibitors of cytochrome P-450, such as the imidazole antifungal agents, also inhibit influx of a calcium surrogate (manganese) into calcium-depleted platelets. Calcium 148-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 8240252-2 1993 This hypothesis is based on the observations that inhibitors of cytochrome P-450, such as the imidazole antifungal agents, also inhibit influx of a calcium surrogate (manganese) into calcium-depleted platelets. Manganese 167-176 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 8257958-4 1993 The increased risk of both H1-antihistamines was associated with exposure to supratherapeutic doses; for terfenadine the risk was also associated with concomitant exposure to the cytochrome P-450 inhibitors ketoconazole, erythromycin and cimetidine. Terfenadine 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-195 8240252-2 1993 This hypothesis is based on the observations that inhibitors of cytochrome P-450, such as the imidazole antifungal agents, also inhibit influx of a calcium surrogate (manganese) into calcium-depleted platelets. Calcium 183-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 8257958-4 1993 The increased risk of both H1-antihistamines was associated with exposure to supratherapeutic doses; for terfenadine the risk was also associated with concomitant exposure to the cytochrome P-450 inhibitors ketoconazole, erythromycin and cimetidine. Cimetidine 238-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-195 8247921-1 1993 Fluconazole is a triazole antifungal agent reported to have a low affinity for human cytochrome P-450, and thus would not be expected to interact with drugs metabolized through the cytochrome P-450 system, including phenytoin, cyclosporine, and warfarin. Fluconazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 7901425-5 1993 These results demonstrate racially distinct patterns of CYP1A1 genotypes, and suggest a functional link between genotype and catalytic activity of the cytochrome P-450 protein responsible for the metabolism of many carcinogenic polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 228-260 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-167 8399332-0 1993 Purification of a human cytochrome P-450 isozyme catalyzing lanosterol 14 alpha-demethylation. Lanosterol 60-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 8399332-1 1993 An isozyme of cytochrome P-450 catalyzing lanosterol 14 alpha-demethylation was purified from human liver using column chromatography, including immunoaffinity chromatography. Lanosterol 42-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 7690968-0 1993 Transfected human liver cytochrome P-450 hydroxylates vitamin D analogs at different side-chain positions. Vitamin D 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 7690968-9 1993 This work has revealed that the cytochrome P-450 CYP27 may be important in the metabolism of vitamin D analogs used as drugs. Vitamin D 93-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 8247921-1 1993 Fluconazole is a triazole antifungal agent reported to have a low affinity for human cytochrome P-450, and thus would not be expected to interact with drugs metabolized through the cytochrome P-450 system, including phenytoin, cyclosporine, and warfarin. Triazoles 17-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 8408733-1 1993 Quinidine and nifedipine appear to be subject to metabolism by the same isozyme of cytochrome P-450. Quinidine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 8415911-4 1993 Of the newly synthesized Pc, two showed significant destruction of cytochrome P-450 and monooxygenase activities, and enhancement of lipid peroxidation, when added to microsomal suspension followed by irradiation with approximately 675 nm light. phthalocyanine 25-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-101 8344930-7 1993 The cytosolic enzyme and the enzymes solubilized from the two pellets produced 12S-hydroperoxy-5,8,10,14-eicosatetraenoic acid as the primary product in contrast to cytochrome P-450 which produces primarily hydroxy acids. Hydroxy Acids 207-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-181 8408733-1 1993 Quinidine and nifedipine appear to be subject to metabolism by the same isozyme of cytochrome P-450. Nifedipine 14-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 8388882-1 1993 Epoxyeicosatrienoic acids (EETs), cytochrome P-450 metabolites of arachidonic acid, have attracted attention because of their effects on stimulus-response coupling in endocrine, renal, and vascular cells. eets 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 8411129-1 1993 Inhibition of the metabolism of arachidonic acid by the epoxygenase (cytochrome P-450) pathway with the inhibitor ketoconazole results in excessive cell swelling upon exposure to hyposmolality instead of the rapid and complete regulatory volume decrease (RVD) normally observed. Arachidonic Acid 32-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 8411129-1 1993 Inhibition of the metabolism of arachidonic acid by the epoxygenase (cytochrome P-450) pathway with the inhibitor ketoconazole results in excessive cell swelling upon exposure to hyposmolality instead of the rapid and complete regulatory volume decrease (RVD) normally observed. Ketoconazole 114-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 7684611-7 1993 These findings suggest that the Ca2+/Mn2+ entry pathway opened by agonists in human neutrophils is the same that activates on emptying the Ca2+ stores and that cytochrome P-450 activity may be involved en the activation of the channels. Manganese(2+) 37-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 8388882-1 1993 Epoxyeicosatrienoic acids (EETs), cytochrome P-450 metabolites of arachidonic acid, have attracted attention because of their effects on stimulus-response coupling in endocrine, renal, and vascular cells. Arachidonic Acid 66-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-50 8388882-11 1993 These findings suggest that hCG stimulates granulosa cell production of EETs via an NADPH-supported, cytochrome P-450-dependent enzymatic mechanism. NADP 84-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-117 7940601-1 1993 The excretion of D-glucaric acid in the urine (uGA) correlates with the total liver content of hepatic cytochrome P-450, the metabolism of which depends on adenosine triphosphate (ATP) being produced by intrahepatic cellular mitochondria. Glucaric Acid 17-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 8332592-4 1993 The first two trajectories were calculated for cytochrome P450cam with thiocamphor bound in both its major and minor crystallographic orientations. thiocamphor 71-82 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 7940601-1 1993 The excretion of D-glucaric acid in the urine (uGA) correlates with the total liver content of hepatic cytochrome P-450, the metabolism of which depends on adenosine triphosphate (ATP) being produced by intrahepatic cellular mitochondria. Adenosine Triphosphate 156-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 7940601-1 1993 The excretion of D-glucaric acid in the urine (uGA) correlates with the total liver content of hepatic cytochrome P-450, the metabolism of which depends on adenosine triphosphate (ATP) being produced by intrahepatic cellular mitochondria. Adenosine Triphosphate 180-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 8490024-0 1993 Regioselectivity of cytochrome P-450 catalyzed hydroxylation of fluorobenzenes predicted by calculated frontier orbital substrate characteristics. Fluorobenzenes 64-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 8389204-0 1993 Omega-hydroxylation of lipoxin B4 by human neutrophil microsomes: identification of omega-hydroxy metabolite of lipoxin B4 and catalysis by leukotriene B4 omega-hydroxylase (cytochrome P-450LTB omega). lipoxin B4 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-172 8389204-0 1993 Omega-hydroxylation of lipoxin B4 by human neutrophil microsomes: identification of omega-hydroxy metabolite of lipoxin B4 and catalysis by leukotriene B4 omega-hydroxylase (cytochrome P-450LTB omega). lipoxin B4 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-199 8389204-4 1993 The reaction is inhibited by carbon monoxide, an inhibitor of cytochrome P-450 (P-450), and by antibodies raised against NADPH-P-450 reductase. Carbon Monoxide 29-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 8389204-6 1993 The P-450 appears to be the one responsible for leukotriene B4 (LTB4) omega-hydroxylation, P-450LTB omega, based on the following observations. Leukotriene B4 48-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-105 8389204-7 1993 The formation of 20-OH-LXB4 is inhibited solely by substrates of P-450LTB omega such as LTB4 and leukotriene B5 among various fatty acids including prostaglandins. 20-Hydroxylipoxin B4 17-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-79 8389204-7 1993 The formation of 20-OH-LXB4 is inhibited solely by substrates of P-450LTB omega such as LTB4 and leukotriene B5 among various fatty acids including prostaglandins. leukotriene B5 97-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-79 8507205-0 1993 The catalytic activity of four expressed human cytochrome P450s towards benzo[a]pyrene and the isomers of its proximate carcinogen. Benzo(a)pyrene 72-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-62 8490024-1 1993 In the present study, a hypothesis is presented for the prediction of the regioselectivity of cytochrome P-450 catalyzed hydroxylation of fluorobenzenes. Fluorobenzenes 138-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 8490024-5 1993 Additional in vitro experiments using 1,2-difluorobenzene as the model substrate demonstrated that minor factors influencing the regioselectivity and possibly responsible for the 6% deviation from the calculated values in in vivo experiments might be (i) the influence of biotransformation routes occurring in vivo but not of importance in in vitro microsomal incubations and (ii) a small variation due to influences of the contribution of various cytochrome P-450 enzymes. 1,2-difluorobenzene 38-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 448-464 8463342-0 1993 Cytochrome P-450 55A1 (P-450dNIR) acts as nitric oxide reductase employing NADH as the direct electron donor. NAD 75-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8100494-5 1993 The rates of formation of 1 and 2 were both correlated with rates of nifedipine oxidation (a marker of cytochrome P-450 (P-450) 3A4) but not with markers for other human P-450s. Nifedipine 69-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 21573617-0 1993 Activation of thio-tepa cytotoxicity toward human breast-cancer cells by hepatic cytochrome-p450. Thiotepa 14-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-96 8291131-0 1993 [Reduction of cytochrome p-450 content and activity by porphyrins after damage to the microsomal membrane by tetrachloromethane]. Porphyrins 55-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 8291131-0 1993 [Reduction of cytochrome p-450 content and activity by porphyrins after damage to the microsomal membrane by tetrachloromethane]. Carbon Tetrachloride 109-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 8291131-1 1993 Natural porphyrin (hemin and its synthetic analogue--tetraphenylporphyrin with iron TPP-Fe3+) has been studied for its effect on activity of monooxygenase responses and cytochrome P-450 content against a background of introduction of four-chlorine carbon in the in vitro experiments. Porphyrins 8-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 8291131-1 1993 Natural porphyrin (hemin and its synthetic analogue--tetraphenylporphyrin with iron TPP-Fe3+) has been studied for its effect on activity of monooxygenase responses and cytochrome P-450 content against a background of introduction of four-chlorine carbon in the in vitro experiments. Hemin 19-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 8291131-1 1993 Natural porphyrin (hemin and its synthetic analogue--tetraphenylporphyrin with iron TPP-Fe3+) has been studied for its effect on activity of monooxygenase responses and cytochrome P-450 content against a background of introduction of four-chlorine carbon in the in vitro experiments. tetraphenylporphyrin 53-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 8291131-2 1993 The reducing effect of hemin and TPP-Fe3+ on cytochrome P-450 and dependent enzymes: N-demethylase, n-hydroxylase and benz(a)pyrene hydroxylase has been noticed; the synthetic analogue of heme was more efficient. Hemin 23-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 8291131-2 1993 The reducing effect of hemin and TPP-Fe3+ on cytochrome P-450 and dependent enzymes: N-demethylase, n-hydroxylase and benz(a)pyrene hydroxylase has been noticed; the synthetic analogue of heme was more efficient. tpp-fe3+ 33-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 8291131-2 1993 The reducing effect of hemin and TPP-Fe3+ on cytochrome P-450 and dependent enzymes: N-demethylase, n-hydroxylase and benz(a)pyrene hydroxylase has been noticed; the synthetic analogue of heme was more efficient. Heme 188-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-61 8486631-5 1993 The microsomes from yeast cells transfected with an expression vector pAAH5 carrying isolated cDNA catalyzed the omega-hydroxylation of LTB4 with a Km value of 0.71 microM, and its activity was significantly inhibited by carbon monoxide and by antisera against CYP4A4, consistent with the properties previously reported with LTB4 omega-hydroxylase in human polymorphonuclear leukocytes. Carbon Monoxide 221-236 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 325-347 8502233-4 1993 Correlations were obtained between rates of imipramine demethylation and cytochrome P-450 (P-450) 1A2 (r = 0.88, p < 0.001) and P-450 3A (r = 0.80, p < 0.02) concentrations in human liver microsomal preparations from 13 different subjects. Imipramine 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 8516280-3 1993 PC and PG liposomes were also treated with PLA2 in the presence of a number of proteins such as cytochrome c, cytochrome b5, cytochrome P-450, polylysine, and histone H1. Phosphatidylglycerols 7-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-169 8387283-5 1993 The unusual substituent effects were also observed in the hydroxylation of p-substituted phenol by microsomes, a cytochrome P-450-dependent reaction. p-substituted phenol 75-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 8463342-2 1993 The cytochrome P-450 (P-450) exhibited a potent nitric oxide (NO) reductase activity to form nitrous oxide (N2O) employing NADH but not NADPH as the sole effective electron donor. Nitric Oxide 48-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 8463342-2 1993 The cytochrome P-450 (P-450) exhibited a potent nitric oxide (NO) reductase activity to form nitrous oxide (N2O) employing NADH but not NADPH as the sole effective electron donor. Nitrous Oxide 93-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 8463342-2 1993 The cytochrome P-450 (P-450) exhibited a potent nitric oxide (NO) reductase activity to form nitrous oxide (N2O) employing NADH but not NADPH as the sole effective electron donor. Nitrous Oxide 108-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 8463342-2 1993 The cytochrome P-450 (P-450) exhibited a potent nitric oxide (NO) reductase activity to form nitrous oxide (N2O) employing NADH but not NADPH as the sole effective electron donor. NAD 123-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 8463342-2 1993 The cytochrome P-450 (P-450) exhibited a potent nitric oxide (NO) reductase activity to form nitrous oxide (N2O) employing NADH but not NADPH as the sole effective electron donor. NADP 136-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 8470252-0 1993 Role of cytochrome P-450 in modulating cyclosporine levels in transplant patients. Cyclosporine 39-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 8386241-3 1993 In contrast, flutamide was oxidatively transformed by cytochrome P-450 into reactive metabolite(s) that covalently bound to microsomal proteins. Flutamide 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 8386241-4 1993 Covalent binding required oxygen and NADPH, and was decreased by the nucleophile glutathione and by the cytochrome P-450 inhibitors SKF 525-A, piperonyl butoxide and troleandomycin (an inhibitor of the cytochrome P-450 3A subfamily). Oxygen 26-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 8386241-4 1993 Covalent binding required oxygen and NADPH, and was decreased by the nucleophile glutathione and by the cytochrome P-450 inhibitors SKF 525-A, piperonyl butoxide and troleandomycin (an inhibitor of the cytochrome P-450 3A subfamily). Oxygen 26-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 8386241-4 1993 Covalent binding required oxygen and NADPH, and was decreased by the nucleophile glutathione and by the cytochrome P-450 inhibitors SKF 525-A, piperonyl butoxide and troleandomycin (an inhibitor of the cytochrome P-450 3A subfamily). NADP 37-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 8386241-4 1993 Covalent binding required oxygen and NADPH, and was decreased by the nucleophile glutathione and by the cytochrome P-450 inhibitors SKF 525-A, piperonyl butoxide and troleandomycin (an inhibitor of the cytochrome P-450 3A subfamily). Glutathione 81-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 8386241-4 1993 Covalent binding required oxygen and NADPH, and was decreased by the nucleophile glutathione and by the cytochrome P-450 inhibitors SKF 525-A, piperonyl butoxide and troleandomycin (an inhibitor of the cytochrome P-450 3A subfamily). Proadifen 132-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 8386241-4 1993 Covalent binding required oxygen and NADPH, and was decreased by the nucleophile glutathione and by the cytochrome P-450 inhibitors SKF 525-A, piperonyl butoxide and troleandomycin (an inhibitor of the cytochrome P-450 3A subfamily). Piperonyl Butoxide 143-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 8386241-4 1993 Covalent binding required oxygen and NADPH, and was decreased by the nucleophile glutathione and by the cytochrome P-450 inhibitors SKF 525-A, piperonyl butoxide and troleandomycin (an inhibitor of the cytochrome P-450 3A subfamily). Piperonyl Butoxide 143-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 8386241-4 1993 Covalent binding required oxygen and NADPH, and was decreased by the nucleophile glutathione and by the cytochrome P-450 inhibitors SKF 525-A, piperonyl butoxide and troleandomycin (an inhibitor of the cytochrome P-450 3A subfamily). Troleandomycin 166-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 104-120 8386241-4 1993 Covalent binding required oxygen and NADPH, and was decreased by the nucleophile glutathione and by the cytochrome P-450 inhibitors SKF 525-A, piperonyl butoxide and troleandomycin (an inhibitor of the cytochrome P-450 3A subfamily). Troleandomycin 166-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 8386241-5 1993 Covalent binding was increased markedly by pretreatment with dexamethasone (an inducer of the cytochrome P-450 3A subfamily) and moderately by pretreatment with beta-naphthoflavone (an inducer of the 1A family). Dexamethasone 61-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 8386241-5 1993 Covalent binding was increased markedly by pretreatment with dexamethasone (an inducer of the cytochrome P-450 3A subfamily) and moderately by pretreatment with beta-naphthoflavone (an inducer of the 1A family). beta-Naphthoflavone 161-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 8463225-1 1993 A cDNA encoding a naturally occurring variant of cytochrome P450 (P450) 2C3 that catalyzes the 6 beta- and 16 alpha-hydroxylation of progesterone exhibits six differences of nucleotide sequence leading to five amino acid substitutions from that encoding 2C3, a progesterone 16 alpha-hydroxylase that does not catalyze 6 beta-hydroxylation. Progesterone 133-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-75 8470644-6 1993 Isoniazid induces the cytochrome P-450 system, resulting in increased metabolism of acetaminophen, formation of toxic metabolites, depletion of glutathione stores, and subsequent hepatocellular injury. Isoniazid 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 8470644-6 1993 Isoniazid induces the cytochrome P-450 system, resulting in increased metabolism of acetaminophen, formation of toxic metabolites, depletion of glutathione stores, and subsequent hepatocellular injury. Acetaminophen 84-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 8470644-6 1993 Isoniazid induces the cytochrome P-450 system, resulting in increased metabolism of acetaminophen, formation of toxic metabolites, depletion of glutathione stores, and subsequent hepatocellular injury. Glutathione 144-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 8470644-7 1993 Patients on isoniazid should use caution when taking acetaminophen since the potentially hepatotoxic effects may be amplified due to induction of the cytochrome P-450 system. Isoniazid 12-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 8470644-7 1993 Patients on isoniazid should use caution when taking acetaminophen since the potentially hepatotoxic effects may be amplified due to induction of the cytochrome P-450 system. Acetaminophen 53-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 150-166 8394802-5 1993 Much has been learned about the mechanism of dioxin"s effects, especially for the induction of cytochrome P-450 enzymes. Dioxins 45-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 8383004-0 1993 Lipid hydroperoxides greatly increase the rate of oxidative catabolism of all-trans-retinoic acid by human cell culture microsomes genetically enriched in specified cytochrome P-450 isoforms. Hydrogen Peroxide 6-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-181 8383004-0 1993 Lipid hydroperoxides greatly increase the rate of oxidative catabolism of all-trans-retinoic acid by human cell culture microsomes genetically enriched in specified cytochrome P-450 isoforms. Tretinoin 74-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-181 8383004-1 1993 Cytochrome P-450 enzymes have been implicated in the oxidative catabolism of all-trans-retinoic acid (RA), a process that is accelerated by exposure to RA in cultured cells and rodents, and also in patients receiving RA as treatment for cancer (J.F.R. Tretinoin 80-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8383004-1 1993 Cytochrome P-450 enzymes have been implicated in the oxidative catabolism of all-trans-retinoic acid (RA), a process that is accelerated by exposure to RA in cultured cells and rodents, and also in patients receiving RA as treatment for cancer (J.F.R. Tretinoin 102-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8383004-1 1993 Cytochrome P-450 enzymes have been implicated in the oxidative catabolism of all-trans-retinoic acid (RA), a process that is accelerated by exposure to RA in cultured cells and rodents, and also in patients receiving RA as treatment for cancer (J.F.R. Tretinoin 152-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8383004-1 1993 Cytochrome P-450 enzymes have been implicated in the oxidative catabolism of all-trans-retinoic acid (RA), a process that is accelerated by exposure to RA in cultured cells and rodents, and also in patients receiving RA as treatment for cancer (J.F.R. Tretinoin 152-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 8452565-0 1993 Cytochrome P450 isoforms in human fetal tissues related to phenobarbital-inducible forms in the mouse. Phenobarbital 59-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 8444412-4 1993 Purification and subfractionation of microsomal protein yielded a fraction of cytochrome P-450, which required NADPH and NADPH-cytochrome P-450 reductase and catalyzed 7 alpha-hydroxylation of the side-chain oxygenated 3 beta-hydroxy-delta 5-C27-steroids but was inactive toward cholesterol. NADP 111-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 8444412-4 1993 Purification and subfractionation of microsomal protein yielded a fraction of cytochrome P-450, which required NADPH and NADPH-cytochrome P-450 reductase and catalyzed 7 alpha-hydroxylation of the side-chain oxygenated 3 beta-hydroxy-delta 5-C27-steroids but was inactive toward cholesterol. Steroids 246-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 8444412-4 1993 Purification and subfractionation of microsomal protein yielded a fraction of cytochrome P-450, which required NADPH and NADPH-cytochrome P-450 reductase and catalyzed 7 alpha-hydroxylation of the side-chain oxygenated 3 beta-hydroxy-delta 5-C27-steroids but was inactive toward cholesterol. Cholesterol 279-290 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 8395243-1 1993 A high affinity binding site for 14(R),15(S)-EET, one of the major cytochrome P-450 metabolites of arachidonic acid (AA) in blood vessels, liver, kidney and urine of patients with pregnancy-induced hypertension, has been identified in a membrane preparation from guinea pig mononuclear (GPM) cells. Arachidonic Acid 99-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-83 8334787-0 1993 [Sparteine oxidation by hepatic cytochrome P-450 in patients with Parkinson"s disease]. Sparteine 1-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 8480473-2 1993 Aldosterone synthase cytochrome P-450 (human P-450aldo) was detected in the tumour portion of aldosterone-producing adenoma, but not in the normal control adrenals, at the protein level. Aldosterone 94-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 8431474-2 1993 The enzymatic activity of bovine adrenocortical cytochrome P-450(11) beta was inhibited by methyltrienolone in a competitive manner without exposure to light and cytochrome P-450(11) beta was photolabeled with methyltrienolone after irradiation with UV light. Metribolone 91-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 8431474-2 1993 The enzymatic activity of bovine adrenocortical cytochrome P-450(11) beta was inhibited by methyltrienolone in a competitive manner without exposure to light and cytochrome P-450(11) beta was photolabeled with methyltrienolone after irradiation with UV light. Metribolone 210-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 8431474-3 1993 The addition of 11-deoxycorticosterone during photolabeling protected cytochrome P-450(11) beta from photolabeling. Desoxycorticosterone 16-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 7922394-0 1993 Prostaglandins, thromboxanes, leukotrienes, and cytochrome P-450 metabolites of arachidonic acid. Arachidonic Acid 80-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 8095148-2 1993 Cytochrome P450 debrisoquine (CYP2D6) activity is polymorphic and under genetic control. Debrisoquin 16-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 7922394-1 1993 Arachidonic acid is metabolized via three major enzymatic pathways: cyclooxygenase, lipoxygenase, and cytochrome P-450. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-118 7922394-4 1993 The cytochrome P-450 pathway metabolites are oxygenated metabolites of arachidonic acid. Arachidonic Acid 71-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 8422907-4 1993 The LXA4 omega-hydroxylation requires both molecular oxygen and NADPH, and is inhibited by carbon monoxide, by antibodies raised against NADPH-cytochrome P-450 reductase, or competitively by leukotriene B4 (LTB4) and LTB5, substrates of LTB4 omega-hydroxylase. Oxygen 53-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 237-259 8454228-2 1993 Debrisoquine/sparteine and mephenytoin polymorphisms are now known to be derived from defects in the human liver of specific forms of microsomal CYP (P450 or previously termed cytochrome P-450), CYP2D6 and CYP2C9 (2C18), respectively. Debrisoquin 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-192 8454228-2 1993 Debrisoquine/sparteine and mephenytoin polymorphisms are now known to be derived from defects in the human liver of specific forms of microsomal CYP (P450 or previously termed cytochrome P-450), CYP2D6 and CYP2C9 (2C18), respectively. Sparteine 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 8454228-2 1993 Debrisoquine/sparteine and mephenytoin polymorphisms are now known to be derived from defects in the human liver of specific forms of microsomal CYP (P450 or previously termed cytochrome P-450), CYP2D6 and CYP2C9 (2C18), respectively. Sparteine 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-192 8454228-2 1993 Debrisoquine/sparteine and mephenytoin polymorphisms are now known to be derived from defects in the human liver of specific forms of microsomal CYP (P450 or previously termed cytochrome P-450), CYP2D6 and CYP2C9 (2C18), respectively. Mephenytoin 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 8454228-2 1993 Debrisoquine/sparteine and mephenytoin polymorphisms are now known to be derived from defects in the human liver of specific forms of microsomal CYP (P450 or previously termed cytochrome P-450), CYP2D6 and CYP2C9 (2C18), respectively. Mephenytoin 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-192 8454228-2 1993 Debrisoquine/sparteine and mephenytoin polymorphisms are now known to be derived from defects in the human liver of specific forms of microsomal CYP (P450 or previously termed cytochrome P-450), CYP2D6 and CYP2C9 (2C18), respectively. Debrisoquin 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 145-148 8475046-1 1993 Site-directed mutants were constructed in cytochrome P-450cam to re-engineer the stereochemistry and coupling of ethylbenzene hydroxylation. ethylbenzene 113-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 8424795-1 1993 Evidence from human studies in vivo and in vitro strongly suggests that the methylhydroxylation of tolbutamide and the 4-hydroxylation of phenytoin, the major pathways in the elimination of these two drugs, are catalysed by the same cytochrome P-450 isoenzyme(s). Tolbutamide 99-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 233-249 8424795-1 1993 Evidence from human studies in vivo and in vitro strongly suggests that the methylhydroxylation of tolbutamide and the 4-hydroxylation of phenytoin, the major pathways in the elimination of these two drugs, are catalysed by the same cytochrome P-450 isoenzyme(s). Phenytoin 138-147 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 233-249 8424795-3 1993 Tolbutamide was hydroxylated to varying extents by all expressed cytochrome P-450 isoenzymes, although activity was much lower for the expressed 2C8 protein. Tolbutamide 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 8422907-4 1993 The LXA4 omega-hydroxylation requires both molecular oxygen and NADPH, and is inhibited by carbon monoxide, by antibodies raised against NADPH-cytochrome P-450 reductase, or competitively by leukotriene B4 (LTB4) and LTB5, substrates of LTB4 omega-hydroxylase. NADP 64-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 237-259 8422907-4 1993 The LXA4 omega-hydroxylation requires both molecular oxygen and NADPH, and is inhibited by carbon monoxide, by antibodies raised against NADPH-cytochrome P-450 reductase, or competitively by leukotriene B4 (LTB4) and LTB5, substrates of LTB4 omega-hydroxylase. Carbon Monoxide 91-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 237-259 8422907-5 1993 These findings indicate that the formation of 20-hydroxy-LXA4 is catalyzed by a neutrophil cytochrome P-450, the LTB4 omega-hydroxylase. 20-hydroxy-lxa4 46-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 8422907-5 1993 These findings indicate that the formation of 20-hydroxy-LXA4 is catalyzed by a neutrophil cytochrome P-450, the LTB4 omega-hydroxylase. 20-hydroxy-lxa4 46-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-135 8445979-0 1993 Amiodarone N-deethylation in human liver microsomes: involvement of cytochrome P450 3A enzymes (first report). Amiodarone 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-83 7922169-5 1993 New information has also become available about the renal biology of cytochrome P-450 metabolites of arachidonic acid as well as about nonenzymatically generated biologically potent eicosanoids, the F2-isoprostanes. Arachidonic Acid 101-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 8070855-0 1993 Characterization of the human cytochrome P450 isozymes responsible for styrene metabolism. Styrene 71-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 8070855-3 1993 The forms of cytochrome P450 that are responsible for transforming styrene to styrene glycol were determined by vaccinia virus-mediated cDNA expression of individual P450 forms in cultured cells. Styrene 67-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 8070855-3 1993 The forms of cytochrome P450 that are responsible for transforming styrene to styrene glycol were determined by vaccinia virus-mediated cDNA expression of individual P450 forms in cultured cells. styrene glycol 78-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 8447722-6 1993 A participation of those isoenzymes of the cytochrome P-450 enzyme system which can be induced by phenobarbital is assumed. Phenobarbital 98-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 8518741-4 1993 The induction process is concomitant with that of cytochrome P-450-IVA1 and cytosolic epoxide hydrolase, which, like bilirubin UGT, are mainly involved in the metabolism of endogenous substrates. Bilirubin 117-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-71 8449148-5 1993 These include myoglobin and hemoglobin, which carry out oxygen binding or transport; mitochondrial cytochromes aa3, b, c, and c3, which are important in transferring electrons; microsomal cytochrome P-450, which catalyzes mixed-function oxidations; catalase, which decomposes H2O2; peroxidase, which activates H2O2; and tryptophan pyrrolase, which catalyzes the oxidation of tryptophan. Hydrogen Peroxide 276-280 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-204 8449148-5 1993 These include myoglobin and hemoglobin, which carry out oxygen binding or transport; mitochondrial cytochromes aa3, b, c, and c3, which are important in transferring electrons; microsomal cytochrome P-450, which catalyzes mixed-function oxidations; catalase, which decomposes H2O2; peroxidase, which activates H2O2; and tryptophan pyrrolase, which catalyzes the oxidation of tryptophan. Hydrogen Peroxide 310-314 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-204 8449148-5 1993 These include myoglobin and hemoglobin, which carry out oxygen binding or transport; mitochondrial cytochromes aa3, b, c, and c3, which are important in transferring electrons; microsomal cytochrome P-450, which catalyzes mixed-function oxidations; catalase, which decomposes H2O2; peroxidase, which activates H2O2; and tryptophan pyrrolase, which catalyzes the oxidation of tryptophan. Tryptophan 320-330 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 188-204 8453968-1 1993 Chlormethiazole is a strong inhibitor of cytochrome P-450-dependent monooxygenases in isolated human liver microsomes. Chlormethiazole 0-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 8445979-1 1993 Experiments were conducted on three different human liver samples to identify the cytochrome P450 isozyme which is involved in the biotransformation of the class III antiarrhythmic agent, amiodarone, into its major metabolite, desethylamiodarone (DEA). Amiodarone 188-198 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 8445979-1 1993 Experiments were conducted on three different human liver samples to identify the cytochrome P450 isozyme which is involved in the biotransformation of the class III antiarrhythmic agent, amiodarone, into its major metabolite, desethylamiodarone (DEA). desethylamiodarone 227-245 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 8445979-1 1993 Experiments were conducted on three different human liver samples to identify the cytochrome P450 isozyme which is involved in the biotransformation of the class III antiarrhythmic agent, amiodarone, into its major metabolite, desethylamiodarone (DEA). desethylamiodarone 247-250 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-97 1461639-13 1992 Chronic ethanol consumption results also in the microsomal induction of a specific ethanol-inducible form of cytochrome P--450, the cytochrome P--450IIE1 with high affinity not only to ethanol but also to some drugs (acetaminophen), procarcinogens (nitrosamines) and industrial agents (carbon tetrachloride). Ethanol 8-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-126 8159952-3 1993 After acute in vivo administration indium and thallium have been shown to produce decreases in the activity of some drug-metabolizing enzymes dependent on cytochrome P-450; therefore these metals would be capable of interfering with the metabolism of organic carcinogens. Indium 35-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 8159952-3 1993 After acute in vivo administration indium and thallium have been shown to produce decreases in the activity of some drug-metabolizing enzymes dependent on cytochrome P-450; therefore these metals would be capable of interfering with the metabolism of organic carcinogens. Thallium 46-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 1472505-6 1992 Coexistence of a mobile population with an average rotational relaxation time phi of 138-577 microseconds and immobile (phi > or = 20 ms) populations of cytochrome P-450 was observed in both phospholipid vesicles and microsomes. Phospholipids 194-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 156-172 1458471-1 1992 The growth-inhibitory effects of ketoconazole, an antifungal agent which inhibits arachidonic acid lipoxygenases and cytochrome P-450 enzymes, were tested in human colon and breast cancer cell lines. Ketoconazole 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 1458471-7 1992 Conversely, clotrimazole and SKF525A, inhibitors of cytochrome P-450 enzymes, had effects similar to those of ketoconazole on HT29-S-B6 cells whereas metronidazole and secnidazole, other azole derivatives which do not inhibit cytochrome P-450 enzymes, had no effect. Clotrimazole 12-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 1458471-7 1992 Conversely, clotrimazole and SKF525A, inhibitors of cytochrome P-450 enzymes, had effects similar to those of ketoconazole on HT29-S-B6 cells whereas metronidazole and secnidazole, other azole derivatives which do not inhibit cytochrome P-450 enzymes, had no effect. Proadifen 29-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 1458471-7 1992 Conversely, clotrimazole and SKF525A, inhibitors of cytochrome P-450 enzymes, had effects similar to those of ketoconazole on HT29-S-B6 cells whereas metronidazole and secnidazole, other azole derivatives which do not inhibit cytochrome P-450 enzymes, had no effect. Proadifen 29-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 226-242 1458471-7 1992 Conversely, clotrimazole and SKF525A, inhibitors of cytochrome P-450 enzymes, had effects similar to those of ketoconazole on HT29-S-B6 cells whereas metronidazole and secnidazole, other azole derivatives which do not inhibit cytochrome P-450 enzymes, had no effect. Azoles 19-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 1458471-8 1992 The results suggest that cytochrome P-450 enzyme(s) activity(ies) could be implicated in the antiproliferative effects of ketoconazole. Ketoconazole 122-134 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 1461639-13 1992 Chronic ethanol consumption results also in the microsomal induction of a specific ethanol-inducible form of cytochrome P--450, the cytochrome P--450IIE1 with high affinity not only to ethanol but also to some drugs (acetaminophen), procarcinogens (nitrosamines) and industrial agents (carbon tetrachloride). Ethanol 83-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-126 1461639-13 1992 Chronic ethanol consumption results also in the microsomal induction of a specific ethanol-inducible form of cytochrome P--450, the cytochrome P--450IIE1 with high affinity not only to ethanol but also to some drugs (acetaminophen), procarcinogens (nitrosamines) and industrial agents (carbon tetrachloride). Ethanol 83-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-126 1461639-13 1992 Chronic ethanol consumption results also in the microsomal induction of a specific ethanol-inducible form of cytochrome P--450, the cytochrome P--450IIE1 with high affinity not only to ethanol but also to some drugs (acetaminophen), procarcinogens (nitrosamines) and industrial agents (carbon tetrachloride). Acetaminophen 217-230 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-126 1461639-13 1992 Chronic ethanol consumption results also in the microsomal induction of a specific ethanol-inducible form of cytochrome P--450, the cytochrome P--450IIE1 with high affinity not only to ethanol but also to some drugs (acetaminophen), procarcinogens (nitrosamines) and industrial agents (carbon tetrachloride). Nitrosamines 249-261 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-126 1461639-13 1992 Chronic ethanol consumption results also in the microsomal induction of a specific ethanol-inducible form of cytochrome P--450, the cytochrome P--450IIE1 with high affinity not only to ethanol but also to some drugs (acetaminophen), procarcinogens (nitrosamines) and industrial agents (carbon tetrachloride). Carbon Tetrachloride 286-306 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-126 1335218-3 1992 There is a strong positive correlation between the rates of ethanol metabolism and the total cytochrome P-450 levels in the hepatoma cells. Ethanol 60-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 1335218-4 1992 The involvement of the cytochrome P-450 system was further supported by the induction of aniline p-hydroxylase activity after ethanol treatment. Ethanol 126-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 1493081-8 1992 The results are consistent with the involvement of the same cytochrome P-450 isoenzyme(s) in the metabolism of tolbutamide and phenytoin. Phenytoin 127-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 1493081-8 1992 The results are consistent with the involvement of the same cytochrome P-450 isoenzyme(s) in the metabolism of tolbutamide and phenytoin. Tolbutamide 111-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 1361312-3 1992 DMED, a 4(5)substituted imidazole, has been shown to inhibit adrenal steroidogenesis and human liver microsomal alfentanil metabolism, reactions mediated by cytochrome P-450. dmed 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 1361312-3 1992 DMED, a 4(5)substituted imidazole, has been shown to inhibit adrenal steroidogenesis and human liver microsomal alfentanil metabolism, reactions mediated by cytochrome P-450. 4(5)substituted imidazole 8-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 1361312-3 1992 DMED, a 4(5)substituted imidazole, has been shown to inhibit adrenal steroidogenesis and human liver microsomal alfentanil metabolism, reactions mediated by cytochrome P-450. Alfentanil 112-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-173 1361312-5 1992 The purpose of this investigation was to determine the mechanism of DMED inhibition of human cytochrome P-450-mediated microsomal metabolism, using ketamine as a probe. Ketamine 148-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 1361312-12 1992 Spectral studies showed that DMED interacted with microsomal cytochrome P-450 to elicit a Type II binding spectrum. dmed 29-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 1475793-6 1992 Under the same experimental conditions, microsomal cytochrome P-450 content was decreased by 40%, 2 h after iron treatment. Iron 108-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 1491046-1 1992 The preparation of samples for injection into a high-performance liquid chromatograph from assay mixtures for the determination of cytochrome P-450-dependent testosterone hydroxylation has been substantially facilitated. Testosterone 158-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 131-147 1426259-0 1992 A critical role of protein-bound water in the catalytic cycle of cytochrome P-450 camphor. Water 33-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 1426259-0 1992 A critical role of protein-bound water in the catalytic cycle of cytochrome P-450 camphor. Camphor 82-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 1491046-5 1992 The modified procedure is a straightforward, easy-to-perform method that should greatly facilitate the implementation of the testosterone hydroxylation assay for sharply discriminating between many individual cytochrome P-450 species in routine enzyme diagnostics. Testosterone 125-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 1468127-0 1992 Effect of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its toxic metabolites on the physicochemical property of the liposomal membrane in relation to their cytochrome P-450 inhibition. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 10-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-183 1488918-12 1992 Stabilization of cytochrome P-450 protein and/or mRNA are the main processes of induction by PCN/glucocorticoids and ethanol-type inducers. Pregnenolone Carbonitrile 93-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 1488918-12 1992 Stabilization of cytochrome P-450 protein and/or mRNA are the main processes of induction by PCN/glucocorticoids and ethanol-type inducers. Ethanol 117-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 1336724-1 1992 The main polycyclic aromatic hydrocarbon-inducible cytochrome P450 was studied in lung tissue from 57 lung cancer patients by immunohistochemistry, using a monoclonal antibody (1-7-1) that recognizes P450IA1 and P450IA2 isozymes. Polycyclic Aromatic Hydrocarbons 9-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-66 1400482-5 1992 Immunoblotting studies demonstrated that dietary salt induced marked increases in the concentration of a cytochrome P-450 isoform(s) recognized by polyclonal antibodies raised against human liver cytochrome P-450 2C10 or rat liver cytochrome P-450 2C11. Salts 49-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 1400482-5 1992 Immunoblotting studies demonstrated that dietary salt induced marked increases in the concentration of a cytochrome P-450 isoform(s) recognized by polyclonal antibodies raised against human liver cytochrome P-450 2C10 or rat liver cytochrome P-450 2C11. Salts 49-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 196-212 1425665-0 1992 Heme-pocket-hydration change during the inactivation of cytochrome P-450camphor by hydrostatic pressure. Heme 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 1334809-0 1992 Calcium influx evoked by Ca2+ store depletion in human platelets is more susceptible to cytochrome P-450 inhibitors than receptor-mediated calcium entry. Calcium 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 1334809-7 1992 In apparent agreement with this, Mn2+ influx promoted by TG and tBuBHQ, or by preincubation of cells in Ca(2+)-free medium, was inhibited by the imidazole antimycotics, econazole and miconazole, which inhibit cytochrome P-450 activity. Manganese(2+) 33-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 1334809-7 1992 In apparent agreement with this, Mn2+ influx promoted by TG and tBuBHQ, or by preincubation of cells in Ca(2+)-free medium, was inhibited by the imidazole antimycotics, econazole and miconazole, which inhibit cytochrome P-450 activity. imidazole 145-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 1334809-7 1992 In apparent agreement with this, Mn2+ influx promoted by TG and tBuBHQ, or by preincubation of cells in Ca(2+)-free medium, was inhibited by the imidazole antimycotics, econazole and miconazole, which inhibit cytochrome P-450 activity. Econazole 169-178 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 209-225 1468127-0 1992 Effect of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its toxic metabolites on the physicochemical property of the liposomal membrane in relation to their cytochrome P-450 inhibition. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 56-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-183 1291449-0 1992 Comparison of cytochrome P-450- and peroxidase-mediated activations of carcinogenic azo dyes and N-nitrosamines. n-nitrosamines 97-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-46 1385058-0 1992 Cytochrome P-450 3A enzymes are responsible for biotransformation of FK506 and rapamycin in man and rat. Tacrolimus 69-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 1328295-0 1992 Human skin levels of retinoic acid and cytochrome P-450-derived 4-hydroxyretinoic acid after topical application of retinoic acid in vivo compared to concentrations required to stimulate retinoic acid receptor-mediated transcription in vitro. 4-hydroxyretinoic acid 64-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 1328295-0 1992 Human skin levels of retinoic acid and cytochrome P-450-derived 4-hydroxyretinoic acid after topical application of retinoic acid in vivo compared to concentrations required to stimulate retinoic acid receptor-mediated transcription in vitro. Tretinoin 73-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 1328295-1 1992 Metabolism of retinoic acid to a less active metabolite, 4-hydroxyretinoic acid, occurs via cytochrome P-450 isozyme(s). Tretinoin 14-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 1328295-1 1992 Metabolism of retinoic acid to a less active metabolite, 4-hydroxyretinoic acid, occurs via cytochrome P-450 isozyme(s). 4-hydroxyretinoic acid 57-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 1328295-6 1992 This cytochrome P-450 mediated activity was oxygen- and NADPH-dependent and was inhibited 68% by 5 microM ketoconazole (P = 0.0035, n = 8) and 51% by carbon monoxide (P = 0.02, n = 6). Oxygen 44-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 1328295-6 1992 This cytochrome P-450 mediated activity was oxygen- and NADPH-dependent and was inhibited 68% by 5 microM ketoconazole (P = 0.0035, n = 8) and 51% by carbon monoxide (P = 0.02, n = 6). NADP 56-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 1328295-6 1992 This cytochrome P-450 mediated activity was oxygen- and NADPH-dependent and was inhibited 68% by 5 microM ketoconazole (P = 0.0035, n = 8) and 51% by carbon monoxide (P = 0.02, n = 6). Ketoconazole 106-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 1328295-6 1992 This cytochrome P-450 mediated activity was oxygen- and NADPH-dependent and was inhibited 68% by 5 microM ketoconazole (P = 0.0035, n = 8) and 51% by carbon monoxide (P = 0.02, n = 6). Carbon Monoxide 150-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 5-21 1519785-3 1992 It has been shown previously that alfentanil clearance is independent of the polymorphic debrisoquine hydroxylase (P-450 2D6), and it is therefore of interest to identify the human cytochrome P-450 enzymes involved in noralfentanil formation, the primary reaction involved in the oxidative N-dealkylation at the piperidine nitrogen. Alfentanil 34-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 1519785-3 1992 It has been shown previously that alfentanil clearance is independent of the polymorphic debrisoquine hydroxylase (P-450 2D6), and it is therefore of interest to identify the human cytochrome P-450 enzymes involved in noralfentanil formation, the primary reaction involved in the oxidative N-dealkylation at the piperidine nitrogen. noralfentanil 218-231 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 1519785-3 1992 It has been shown previously that alfentanil clearance is independent of the polymorphic debrisoquine hydroxylase (P-450 2D6), and it is therefore of interest to identify the human cytochrome P-450 enzymes involved in noralfentanil formation, the primary reaction involved in the oxidative N-dealkylation at the piperidine nitrogen. piperidine 312-322 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 1519785-3 1992 It has been shown previously that alfentanil clearance is independent of the polymorphic debrisoquine hydroxylase (P-450 2D6), and it is therefore of interest to identify the human cytochrome P-450 enzymes involved in noralfentanil formation, the primary reaction involved in the oxidative N-dealkylation at the piperidine nitrogen. Nitrogen 323-331 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-197 1525169-2 1992 Cytochrome P450 oxidizes T-5-O to the 5,10- rather than the 5,5-dioxide but oxidizes the 5,5-dioxide rapidly and the 5,10-dioxide slowly to the 5,5,10-trioxide. thianthrene 5-oxide 25-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1525169-2 1992 Cytochrome P450 oxidizes T-5-O to the 5,10- rather than the 5,5-dioxide but oxidizes the 5,5-dioxide rapidly and the 5,10-dioxide slowly to the 5,5,10-trioxide. 5,5-dioxide 60-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1525169-2 1992 Cytochrome P450 oxidizes T-5-O to the 5,10- rather than the 5,5-dioxide but oxidizes the 5,5-dioxide rapidly and the 5,10-dioxide slowly to the 5,5,10-trioxide. 5,5-dioxide 89-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1525169-2 1992 Cytochrome P450 oxidizes T-5-O to the 5,10- rather than the 5,5-dioxide but oxidizes the 5,5-dioxide rapidly and the 5,10-dioxide slowly to the 5,5,10-trioxide. 5,10-dioxide 117-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1525169-2 1992 Cytochrome P450 oxidizes T-5-O to the 5,10- rather than the 5,5-dioxide but oxidizes the 5,5-dioxide rapidly and the 5,10-dioxide slowly to the 5,5,10-trioxide. 5,5,10-trioxide 144-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1525169-12 1992 A large difference in the cis:trans ratios of the 5,10-dioxides produced from T-5-O by cytochrome P450 (1.3:1) and chloroperoxidase (2.5:1) vs hemoglobin (0.1:1) suggests that the hemoglobin active site severely constrains the geometry of the electrophilic oxidation. 5,10-dioxides 50-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 1525169-12 1992 A large difference in the cis:trans ratios of the 5,10-dioxides produced from T-5-O by cytochrome P450 (1.3:1) and chloroperoxidase (2.5:1) vs hemoglobin (0.1:1) suggests that the hemoglobin active site severely constrains the geometry of the electrophilic oxidation. thianthrene 5-oxide 78-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 1463937-2 1992 Successive treatment of microsomes with protease XXVII and 3-(3-cholamidopropyl)dimethylammoniopropanesulphonic acid gave a solubilized cyt-P450 in more than 80% yield and with a three-fold increase in specific activity. 3-(3-cholamidopropyl)dimethylammoniopropanesulphonic acid 59-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 136-144 1463937-4 1992 The solubilized cyt-P450 was filtered on a Sephacryl-200 column and then subjected to high performance liquid chromatography with a Mono-P column (chromatofocussing). mono-p 132-138 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-24 1385058-0 1992 Cytochrome P-450 3A enzymes are responsible for biotransformation of FK506 and rapamycin in man and rat. Sirolimus 79-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 1385058-1 1992 The hepatic cytochrome P-450 responsible for metabolism of the structurally related macrolides FK506 and rapamycin in humans was identified using in vitro studies. Macrolides 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 1521541-2 1992 Cytochrome P-450 species (P-450) comprise a polymorphic multigene family of heme-containing enzymes which are essential to the phase-I metabolism of xenobiotics. Heme 76-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 1385058-1 1992 The hepatic cytochrome P-450 responsible for metabolism of the structurally related macrolides FK506 and rapamycin in humans was identified using in vitro studies. Tacrolimus 95-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 1385058-1 1992 The hepatic cytochrome P-450 responsible for metabolism of the structurally related macrolides FK506 and rapamycin in humans was identified using in vitro studies. Sirolimus 105-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 12-28 1385058-7 1992 It is concluded that in human and rat liver FK506 and rapamycin are metabolized primarily by cytochrome P-450 3A4. Tacrolimus 44-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 1385058-7 1992 It is concluded that in human and rat liver FK506 and rapamycin are metabolized primarily by cytochrome P-450 3A4. Sirolimus 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 1457335-0 1992 The renal cytochrome P-450 arachidonic acid system. Arachidonic Acid 27-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 1505911-1 1992 The localization of the expression of several cytochrome P-450 genes in normal and diseased human liver was investigated by in situ hybridization of formalin-fixed, paraffin wax-embedded archival tissue samples with 35S-labeled antisense RNA probes. Formaldehyde 149-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 1505911-1 1992 The localization of the expression of several cytochrome P-450 genes in normal and diseased human liver was investigated by in situ hybridization of formalin-fixed, paraffin wax-embedded archival tissue samples with 35S-labeled antisense RNA probes. Paraffin 165-177 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 1505911-1 1992 The localization of the expression of several cytochrome P-450 genes in normal and diseased human liver was investigated by in situ hybridization of formalin-fixed, paraffin wax-embedded archival tissue samples with 35S-labeled antisense RNA probes. Sulfur-35 216-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 1331779-1 1992 Aromatase, a cytochrome P-450, catalyzes the formation of aromatic C18 estrogenic steroids from C19 androgens. Steroids 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 1457335-3 1992 AA is metabolized by the renal cytochrome P-450 epoxygenase and omega/omega 1 hydroxylases to epoxyeicosatrienoic acids and omega/omega-1 alcohols (20- and 19-mono-hydroxyeicosatetraenoic acids), respectively. epoxyeicosatrienoic acids 94-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 1457335-3 1992 AA is metabolized by the renal cytochrome P-450 epoxygenase and omega/omega 1 hydroxylases to epoxyeicosatrienoic acids and omega/omega-1 alcohols (20- and 19-mono-hydroxyeicosatetraenoic acids), respectively. omega/omega-1 alcohols 124-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 1457335-3 1992 AA is metabolized by the renal cytochrome P-450 epoxygenase and omega/omega 1 hydroxylases to epoxyeicosatrienoic acids and omega/omega-1 alcohols (20- and 19-mono-hydroxyeicosatetraenoic acids), respectively. mono-hydroxyeicosatetraenoic acids 159-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 1441599-0 1992 Polymorphism in stereoselective hydroxylations of mephenytoin and hexobarbital by Japanese liver samples in relation to cytochrome P-450 human-2 (IIC9). Mephenytoin 50-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 1344831-4 1992 Omeprazole interacts with the cytochrome P-450 system in the liver: inhibition of several liver mono-oxygenases activities (inhibitory effect on diazepam, phenytoin and R-warfarin metabolism with prolonged elimination); induction of P-450 (IA1 and IA2) enzymes that may potentiate the hepatotoxic effect of phenacetin and acetaminophen or increase the tumorigenic effect of chemical carcinogens (polycyclic aromatic hydrocarbons, arylamines, aflatoxin). Omeprazole 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 1344831-4 1992 Omeprazole interacts with the cytochrome P-450 system in the liver: inhibition of several liver mono-oxygenases activities (inhibitory effect on diazepam, phenytoin and R-warfarin metabolism with prolonged elimination); induction of P-450 (IA1 and IA2) enzymes that may potentiate the hepatotoxic effect of phenacetin and acetaminophen or increase the tumorigenic effect of chemical carcinogens (polycyclic aromatic hydrocarbons, arylamines, aflatoxin). Diazepam 145-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 1344831-4 1992 Omeprazole interacts with the cytochrome P-450 system in the liver: inhibition of several liver mono-oxygenases activities (inhibitory effect on diazepam, phenytoin and R-warfarin metabolism with prolonged elimination); induction of P-450 (IA1 and IA2) enzymes that may potentiate the hepatotoxic effect of phenacetin and acetaminophen or increase the tumorigenic effect of chemical carcinogens (polycyclic aromatic hydrocarbons, arylamines, aflatoxin). Phenytoin 155-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 1344831-4 1992 Omeprazole interacts with the cytochrome P-450 system in the liver: inhibition of several liver mono-oxygenases activities (inhibitory effect on diazepam, phenytoin and R-warfarin metabolism with prolonged elimination); induction of P-450 (IA1 and IA2) enzymes that may potentiate the hepatotoxic effect of phenacetin and acetaminophen or increase the tumorigenic effect of chemical carcinogens (polycyclic aromatic hydrocarbons, arylamines, aflatoxin). Phenacetin 307-317 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 1344831-4 1992 Omeprazole interacts with the cytochrome P-450 system in the liver: inhibition of several liver mono-oxygenases activities (inhibitory effect on diazepam, phenytoin and R-warfarin metabolism with prolonged elimination); induction of P-450 (IA1 and IA2) enzymes that may potentiate the hepatotoxic effect of phenacetin and acetaminophen or increase the tumorigenic effect of chemical carcinogens (polycyclic aromatic hydrocarbons, arylamines, aflatoxin). Acetaminophen 322-335 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 1344831-4 1992 Omeprazole interacts with the cytochrome P-450 system in the liver: inhibition of several liver mono-oxygenases activities (inhibitory effect on diazepam, phenytoin and R-warfarin metabolism with prolonged elimination); induction of P-450 (IA1 and IA2) enzymes that may potentiate the hepatotoxic effect of phenacetin and acetaminophen or increase the tumorigenic effect of chemical carcinogens (polycyclic aromatic hydrocarbons, arylamines, aflatoxin). Polycyclic Aromatic Hydrocarbons 396-428 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 1344831-4 1992 Omeprazole interacts with the cytochrome P-450 system in the liver: inhibition of several liver mono-oxygenases activities (inhibitory effect on diazepam, phenytoin and R-warfarin metabolism with prolonged elimination); induction of P-450 (IA1 and IA2) enzymes that may potentiate the hepatotoxic effect of phenacetin and acetaminophen or increase the tumorigenic effect of chemical carcinogens (polycyclic aromatic hydrocarbons, arylamines, aflatoxin). aniline 430-440 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 1344831-4 1992 Omeprazole interacts with the cytochrome P-450 system in the liver: inhibition of several liver mono-oxygenases activities (inhibitory effect on diazepam, phenytoin and R-warfarin metabolism with prolonged elimination); induction of P-450 (IA1 and IA2) enzymes that may potentiate the hepatotoxic effect of phenacetin and acetaminophen or increase the tumorigenic effect of chemical carcinogens (polycyclic aromatic hydrocarbons, arylamines, aflatoxin). Aflatoxins 442-451 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 1441599-16 1992 These findings indicate that P-450 human-2 is the major cytochrome P-450 responsible for the polymorphisms in stereoselective hydroxylations of mephenytoin and hexobarbital. Mephenytoin 144-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-72 1441602-2 1992 Microsomal reduction of azo dyes related to dimethylaminoazobenzene (DAB) is catalysed by at least two types of cytochrome P-450. Azo Compounds 24-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 1441602-2 1992 Microsomal reduction of azo dyes related to dimethylaminoazobenzene (DAB) is catalysed by at least two types of cytochrome P-450. p-Dimethylaminoazobenzene 44-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 1441602-2 1992 Microsomal reduction of azo dyes related to dimethylaminoazobenzene (DAB) is catalysed by at least two types of cytochrome P-450. p-Dimethylaminoazobenzene 69-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-128 1504011-3 1992 In addition, it has been found that malondialdehyde formation is accompanied by a corresponding decrease in placental mitochondrial cytochrome P-450 content. Malondialdehyde 36-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-148 1643634-0 1992 Role of cytochrome P-450 from the human CYP3A gene family in the potentiation of morpholino doxorubicin by human liver microsomes. NSC 354646 81-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 1643634-3 1992 This metabolic potentiation is inhibited by carbon monoxide or hypoxia, indicating that it is cytochrome P-450-dependent. Carbon Monoxide 44-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 1643634-4 1992 The potentiation is also inhibited by the cytochrome P-450 inhibitors, SKF-525A and cimetidine. Cimetidine 84-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 1510705-3 1992 Ketoconazole, a known inhibitor of cytochrome P450 isozymes, caused marked inhibition of carboxyprimaquine formation with IC50 and K(i) values of 15 and 6.7 microM, respectively. Ketoconazole 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 1510705-3 1992 Ketoconazole, a known inhibitor of cytochrome P450 isozymes, caused marked inhibition of carboxyprimaquine formation with IC50 and K(i) values of 15 and 6.7 microM, respectively. 8-(3-carboxy-1-methylpropylamino)-6-methoxyquinoline 89-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-50 1510705-4 1992 This finding and the dependency of metabolite formation on NADPH indicates that cytochrome P450 isozyme(s) catalysed metabolite production. NADP 59-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 1617624-8 1992 Metabolite yields under various conditions were determined, including a demonstration that carbon monoxide inhibited 81% of NMAA demethylation, indicating that cytochrome P-450 enzymes were involved. Carbon Monoxide 91-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 1496595-0 1992 Correlation of cyclosporine-induced nephropathy with renal microsomal cytochrome P-450: the preventive effect of verapamil. Cyclosporine 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 1379068-8 1992 The reduced carbon monoxide binding spectrum shows an absorption maximum at 447 nm indicative of a cytochrome P-450 hemoprotein. Carbon Monoxide 12-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 1627548-9 1992 These results suggested that the stimulation of catalytic activity by phospholipids involved increased interaction between cytochrome P-450 and the reductase. Phospholipids 70-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 1617624-8 1992 Metabolite yields under various conditions were determined, including a demonstration that carbon monoxide inhibited 81% of NMAA demethylation, indicating that cytochrome P-450 enzymes were involved. nmaa 124-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 1327375-2 1992 The cytochrome P-450 metabolite of arachidonic acid, 20-hydroxyeicosatetraenoic acid (20-HETE), was found to be a potent, dose-dependent inhibitor of platelet aggregation and inhibitor of thromboxane biosynthesis induced by arachidonic acid (IC50 5.2 +/- 1.5 microM), A23187 (IC50 16.2 +/- 5.4 microM), and U46619 (IC50 7.8 +/- 2.4 microM). Arachidonic Acid 35-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 1628663-0 1992 The cholesterol-side-chain-cleaving cytochrome P450 spin-state equilibrium. Cholesterol 4-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-51 1628663-3 1992 We have confirmed and characterised structurally the enzyme conformational changes deduced from the preceding thermodynamic analysis of the adrenal mitochondrial cholesterol-side-chain-cleaving cytochrome P450 spin-state equilibrium. Cholesterol 162-173 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-209 1524698-3 1992 The main mechanism of most drug interactions involving cimetidine appears to be inhibition of the hepatic microsomal enzyme cytochrome P450, an effect which may be related to the different structures of H2-antagonists. Cimetidine 55-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 1524698-3 1992 The main mechanism of most drug interactions involving cimetidine appears to be inhibition of the hepatic microsomal enzyme cytochrome P450, an effect which may be related to the different structures of H2-antagonists. Hydrogen 203-205 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 1327375-2 1992 The cytochrome P-450 metabolite of arachidonic acid, 20-hydroxyeicosatetraenoic acid (20-HETE), was found to be a potent, dose-dependent inhibitor of platelet aggregation and inhibitor of thromboxane biosynthesis induced by arachidonic acid (IC50 5.2 +/- 1.5 microM), A23187 (IC50 16.2 +/- 5.4 microM), and U46619 (IC50 7.8 +/- 2.4 microM). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 53-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 1327375-2 1992 The cytochrome P-450 metabolite of arachidonic acid, 20-hydroxyeicosatetraenoic acid (20-HETE), was found to be a potent, dose-dependent inhibitor of platelet aggregation and inhibitor of thromboxane biosynthesis induced by arachidonic acid (IC50 5.2 +/- 1.5 microM), A23187 (IC50 16.2 +/- 5.4 microM), and U46619 (IC50 7.8 +/- 2.4 microM). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 86-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 1327375-2 1992 The cytochrome P-450 metabolite of arachidonic acid, 20-hydroxyeicosatetraenoic acid (20-HETE), was found to be a potent, dose-dependent inhibitor of platelet aggregation and inhibitor of thromboxane biosynthesis induced by arachidonic acid (IC50 5.2 +/- 1.5 microM), A23187 (IC50 16.2 +/- 5.4 microM), and U46619 (IC50 7.8 +/- 2.4 microM). Thromboxanes 188-199 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 1327375-2 1992 The cytochrome P-450 metabolite of arachidonic acid, 20-hydroxyeicosatetraenoic acid (20-HETE), was found to be a potent, dose-dependent inhibitor of platelet aggregation and inhibitor of thromboxane biosynthesis induced by arachidonic acid (IC50 5.2 +/- 1.5 microM), A23187 (IC50 16.2 +/- 5.4 microM), and U46619 (IC50 7.8 +/- 2.4 microM). Arachidonic Acid 224-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 1327375-2 1992 The cytochrome P-450 metabolite of arachidonic acid, 20-hydroxyeicosatetraenoic acid (20-HETE), was found to be a potent, dose-dependent inhibitor of platelet aggregation and inhibitor of thromboxane biosynthesis induced by arachidonic acid (IC50 5.2 +/- 1.5 microM), A23187 (IC50 16.2 +/- 5.4 microM), and U46619 (IC50 7.8 +/- 2.4 microM). Calcimycin 268-274 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 1321524-1 1992 Dioxins and peroxisome proliferators represent two diverse classes of xenobiotic compounds that induce transcription of specific genes encoding cytochrome P-450 drug-metabolizing enzymes. Dioxins 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 144-160 1316878-8 1992 This indicates that inhibition of LTB4 omega-hydroxylase activity by pyocyanin might be achieved by competition for NADPH. Pyocyanine 69-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-56 1316878-8 1992 This indicates that inhibition of LTB4 omega-hydroxylase activity by pyocyanin might be achieved by competition for NADPH. NADP 116-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-56 1640215-2 1992 The term cytochrome P450 refers to a group of hemoproteins whose Fe(2+)-carbon monoxide complex shows an absorption spectrum with a maximum near 450 nm. fe(2+)-carbon monoxide 65-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-24 1640215-4 1992 Cytochrome P450 enzymes are involved in the metabolism of drugs, carcinogens, steroids, pesticides, hydrocarbons and natural products. Steroids 78-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1640215-4 1992 Cytochrome P450 enzymes are involved in the metabolism of drugs, carcinogens, steroids, pesticides, hydrocarbons and natural products. Hydrocarbons 100-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1596283-5 1992 The inhibition of ketoconazole, a known inhibitor of cytochrome P450 isozymes, and the dependency of metabolite formation on the presence of NADPH indicated that cytochrome P450 isozyme(s) catalysed metabolite production. Ketoconazole 18-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 1587824-0 1992 Reconstitution of the fatty acid hydroxylation function of cytochrome P-450BM-3 utilizing its individual recombinant hemo- and flavoprotein domains. Fatty Acids 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-79 1587824-3 1992 Prior attempts to reconstitute the fatty acid hydroxylation function of cytochrome P-450BM-3, utilizing the two domains, obtained either by trypsinolysis or by recombinant methods, were unsuccessful. Fatty Acids 35-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-92 1587824-4 1992 In this paper, we describe the reconstitution of the fatty acid hydroxylation activity of cytochrome P-450BM-3 utilizing the recombinantly produced flavoprotein domain (Oster, T., Boddupalli, S. S., and Peterson, J. Fatty Acids 53-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-110 1587824-10 1992 The combination of the hemo- and flavoprotein domains in a ratio of 20:1 respectively, in the reaction mixture, results in the transfer of 80% of the reducing equivalents from NADPH for the hydroxylation of palmitate at 25 degrees C. The ratio of the regioisomeric products obtained for lauric, myristic, and palmitic acids was similar to that obtained with the holoenzyme form of cytochrome P-450BM-3. NADP 176-181 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 381-401 1587824-11 1992 The reconstitution of the fatty acid omega-hydroxylase activity, using the soluble domains of cytochrome P-450BM-3, without added factors such as lipids, may be useful for structure/function comparisons to their eukaryotic counterparts. Fatty Acids 26-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-114 1596283-5 1992 The inhibition of ketoconazole, a known inhibitor of cytochrome P450 isozymes, and the dependency of metabolite formation on the presence of NADPH indicated that cytochrome P450 isozyme(s) catalysed metabolite production. Ketoconazole 18-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 1596283-5 1992 The inhibition of ketoconazole, a known inhibitor of cytochrome P450 isozymes, and the dependency of metabolite formation on the presence of NADPH indicated that cytochrome P450 isozyme(s) catalysed metabolite production. NADP 141-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-68 1596283-5 1992 The inhibition of ketoconazole, a known inhibitor of cytochrome P450 isozymes, and the dependency of metabolite formation on the presence of NADPH indicated that cytochrome P450 isozyme(s) catalysed metabolite production. NADP 141-146 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-177 1594575-0 1992 Substrate mobility in a deeply buried active site: analysis of norcamphor bound to cytochrome P-450cam as determined by a 201-psec molecular dynamics simulation. norcamphor 63-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 1423213-2 1992 The increased steroid production demonstrated by most of the pathological tissue samples examined here was associated with either an unchanged or dramatically decreased specific microsomal content of cytochrome P-450. Steroids 14-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-216 1610865-5 1992 Clearance data suggests, however, that the primary mechanism for the interaction is the inhibition cyclosporine metabolism by the cytochrome P-450 system. Cyclosporine 99-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 1394641-0 1992 Fluconazole: a potent inhibitor of cytochrome P-450-dependent drug-metabolism in mice and humans in vivo. Fluconazole 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 1394641-2 1992 The inhibitory effect of fluconazole (FCZ), a bis-triazole antimycotic, on mouse hepatic microsomal cytochrome P-450-mediated drug-metabolizing enzyme system was compared with those of ketoconazole (KCZ) in vivo and in vitro. Fluconazole 25-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 1394641-2 1992 The inhibitory effect of fluconazole (FCZ), a bis-triazole antimycotic, on mouse hepatic microsomal cytochrome P-450-mediated drug-metabolizing enzyme system was compared with those of ketoconazole (KCZ) in vivo and in vitro. Fluconazole 38-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 100-116 1394641-5 1992 In contrast, in vitro the affinity and the inhibitory potency of FCZ for cytochrome P-450 and aminopyrine N-demethylation were 4- to 6-fold smaller than those of KCZ. Fluconazole 65-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 1578365-0 1992 Human cytochrome P450 metabolism of teniposide and etoposide. Teniposide 36-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 1578365-0 1992 Human cytochrome P450 metabolism of teniposide and etoposide. Etoposide 51-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 6-21 1354126-2 1992 Cytochrome P-450 was induced in adult hen liver by administering 15% ethanol in drinking water and compared with other inducers such as phenobarbital and beta-naphthoflavone. Ethanol 69-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 1539561-1 1992 Both cimetidine and cyclosporine (CsA) are metabolized by the cytochrome P-450 system. Cimetidine 5-15 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 1312930-0 1992 Effect of KCl on the interactions between NADPH:cytochrome P-450 reductase and either cytochrome c, cytochrome b5 or cytochrome P-450 in octyl glucoside micelles. Potassium Chloride 10-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 1312930-0 1992 Effect of KCl on the interactions between NADPH:cytochrome P-450 reductase and either cytochrome c, cytochrome b5 or cytochrome P-450 in octyl glucoside micelles. octyl-beta-D-glucoside 137-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 1312930-1 1992 Significant dissociation of FMN from NADPH:cytochrome P-450 reductase resulted in loss of the activity for reduction of cytochrome b5 as well as cytochrome c and cytochrome P-450. Flavin Mononucleotide 28-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 1312930-3 1992 The reductions of cytochrome c and detergent-solubilized cytochrome b5 by NADPH:cytochrome P-450 reductase were greatly increased in the presence of high concentrations of KCl, although the stimulatory effect of the salt on cytochrome P-450 reduction was less significant. Potassium Chloride 172-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 1312930-3 1992 The reductions of cytochrome c and detergent-solubilized cytochrome b5 by NADPH:cytochrome P-450 reductase were greatly increased in the presence of high concentrations of KCl, although the stimulatory effect of the salt on cytochrome P-450 reduction was less significant. Salts 216-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 1523868-11 1992 The increase in toxicity of heroin and methadone produced by ethanol is concomitant with a 40% increase in cytochrome P-450 levels of the pretreated hepatocytes. Heroin 28-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-123 1523868-11 1992 The increase in toxicity of heroin and methadone produced by ethanol is concomitant with a 40% increase in cytochrome P-450 levels of the pretreated hepatocytes. Ethanol 61-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 107-123 1643250-3 1992 Aflatoxin Q1 and aflatoxin B1 8,9-oxide (trapped here as the glutathione conjugate) are the major oxidative products formed from aflatoxin B1 at all substrate concentrations in human liver microsomes, and cytochrome P-450 (P-450) 3A4 appears to be the dominant enzyme involved in both oxidations, as judged by studies involving correlation of activities in different liver samples, chemical inhibition, immunoinhibition, and reconstitution with purified hepatic and yeast recombinant P-450 3A4. aflatoxin Q1 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-221 1643250-3 1992 Aflatoxin Q1 and aflatoxin B1 8,9-oxide (trapped here as the glutathione conjugate) are the major oxidative products formed from aflatoxin B1 at all substrate concentrations in human liver microsomes, and cytochrome P-450 (P-450) 3A4 appears to be the dominant enzyme involved in both oxidations, as judged by studies involving correlation of activities in different liver samples, chemical inhibition, immunoinhibition, and reconstitution with purified hepatic and yeast recombinant P-450 3A4. aflatoxin B1-2,3-oxide 17-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-221 1643250-3 1992 Aflatoxin Q1 and aflatoxin B1 8,9-oxide (trapped here as the glutathione conjugate) are the major oxidative products formed from aflatoxin B1 at all substrate concentrations in human liver microsomes, and cytochrome P-450 (P-450) 3A4 appears to be the dominant enzyme involved in both oxidations, as judged by studies involving correlation of activities in different liver samples, chemical inhibition, immunoinhibition, and reconstitution with purified hepatic and yeast recombinant P-450 3A4. Glutathione 61-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-221 32357628-3 1992 Recent purification and characterization of specific steroidogenic enzymes, such as cytochrome P-450, that are involved in adrenal steroid biosynthesis have made it possible to generate specific antibodies and DNA probes against the enzymes that catalyze specific reaction(s) in the complicated biochemical cascade of steroidogenesis. Steroids 53-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 1539561-1 1992 Both cimetidine and cyclosporine (CsA) are metabolized by the cytochrome P-450 system. Cyclosporine 20-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 1539561-1 1992 Both cimetidine and cyclosporine (CsA) are metabolized by the cytochrome P-450 system. Cyclosporine 34-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 1546013-2 1992 The mechanism of increased susceptibility involves induction of isoenzymes of the cytochrome P-450 system by alcohol and depletion of hepatic glutathione reserves, both of which can result from chronic alcohol ingestion and both of which affect the metabolism of acetaminophen. Alcohols 109-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 1546013-2 1992 The mechanism of increased susceptibility involves induction of isoenzymes of the cytochrome P-450 system by alcohol and depletion of hepatic glutathione reserves, both of which can result from chronic alcohol ingestion and both of which affect the metabolism of acetaminophen. Alcohols 202-209 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 1546013-2 1992 The mechanism of increased susceptibility involves induction of isoenzymes of the cytochrome P-450 system by alcohol and depletion of hepatic glutathione reserves, both of which can result from chronic alcohol ingestion and both of which affect the metabolism of acetaminophen. Acetaminophen 263-276 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 82-98 1737335-0 1992 A new form of cytochrome P-450 responsible for mutagenic activation of 2-amino-3-methylimidazo[4,5-f]quinoline in human livers. 2-amino-3-methylimidazo(4,5-f)quinoline 71-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 14-30 1537463-1 1992 Arachidonic acid and many products of the arachidonate cascade serve as substrates for cytochrome P450-mediated metabolism via allylic oxidation, omega hydroxylation, and epoxygenation as well as peroxide rearrangement. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 1550407-4 1992 The synthesis of 1,25(OH)2D3 and 24,25(OH)2D3 by macrophages and fibroblast-like cells respectively was inhibited by ketoconazole, indicating that both hydroxylases are dependent on cytochrome P-450. Calcitriol 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-198 1550407-4 1992 The synthesis of 1,25(OH)2D3 and 24,25(OH)2D3 by macrophages and fibroblast-like cells respectively was inhibited by ketoconazole, indicating that both hydroxylases are dependent on cytochrome P-450. 24,25(oh)2d3 33-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-198 1550407-4 1992 The synthesis of 1,25(OH)2D3 and 24,25(OH)2D3 by macrophages and fibroblast-like cells respectively was inhibited by ketoconazole, indicating that both hydroxylases are dependent on cytochrome P-450. Ketoconazole 117-129 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-198 1311943-7 1992 These data confirm that ketoconazole is a potent inhibitor of cytochrome P-450-IIIA4, and this has clinical relevance. Ketoconazole 24-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-84 1311944-2 1992 Agents such as substituent imidazoles and triazoles, which act by inhibiting the fungal cytochrome P-450-dependent enzyme lanosterol N-demethylase, have the potential to inhibit host cytochrome P-450-dependent drug metabolism. Imidazoles 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 1311944-2 1992 Agents such as substituent imidazoles and triazoles, which act by inhibiting the fungal cytochrome P-450-dependent enzyme lanosterol N-demethylase, have the potential to inhibit host cytochrome P-450-dependent drug metabolism. Imidazoles 27-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 1311944-2 1992 Agents such as substituent imidazoles and triazoles, which act by inhibiting the fungal cytochrome P-450-dependent enzyme lanosterol N-demethylase, have the potential to inhibit host cytochrome P-450-dependent drug metabolism. Triazoles 42-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 1311944-2 1992 Agents such as substituent imidazoles and triazoles, which act by inhibiting the fungal cytochrome P-450-dependent enzyme lanosterol N-demethylase, have the potential to inhibit host cytochrome P-450-dependent drug metabolism. Triazoles 42-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-199 1311944-4 1992 In contrast, allylamines, which have a different mode of action and a weaker ability to bind to cytochrome P-450, are not expected to inhibit clinical drug oxidation. Allylamine 13-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 1373902-6 1992 Western blot analysis of liver microsomes with antibodies to rat P-450IA2 (P-448-H), P-450IIB1 (P-450b) and human P-450IIIA4 (P-450NF) showed that ascorbic acid-deficiency resulted in a decrease in the amount of cytochrome P-450 immunochemically related to P-450IA2, but not the amounts of the forms of cytochrome P-450 cross-reactive with antibodies to P-450IIB1 and P-450IIIA4. Ascorbic Acid 147-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 303-319 1730667-1 1992 Aromatase, a cytochrome P-450, catalyzes the formation of aromatic C18 estrogenic steroids from C19 androgens. Steroids 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 1539615-4 1992 Two separate studies have shown that Med I"s vasodepressor action is inhibited by four procedures: mixing with Tween 20; treatment with n-butyl boronic acid; treatment of the assay animal with SKF 525A, an inhibitor of cytochrome P-450; and removing the liver from the circulation. Proadifen 193-201 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 219-235 1536639-0 1992 Phenobarbital induction of cytochrome P-450 gene expression. Phenobarbital 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 1537463-1 1992 Arachidonic acid and many products of the arachidonate cascade serve as substrates for cytochrome P450-mediated metabolism via allylic oxidation, omega hydroxylation, and epoxygenation as well as peroxide rearrangement. Arachidonic Acid 42-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-102 1537463-3 1992 Cytochrome P450-catalyzed reactions play prominent roles in multiplying the structural and functional diversity of the arachidonate metabolic cascade. Arachidonic Acid 119-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1537469-3 1992 The acceleration of Mn2+ entry observed in cells with empty intracellular Ca2+ stores was prevented by cytochrome P450 inhibitors. Manganese(2+) 20-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 1581524-0 1992 A new hypothesis for the mechanism for cytochrome P-450 dependent aerobic conversion of hexahalogenated benzenes to pentahalogenated phenols. Benzene 104-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 1581524-0 1992 A new hypothesis for the mechanism for cytochrome P-450 dependent aerobic conversion of hexahalogenated benzenes to pentahalogenated phenols. Phenols 133-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 1581537-0 1992 Hydroxylation of warfarin by human cDNA-expressed cytochrome P-450: a role for P-4502C9 in the etiology of (S)-warfarin-drug interactions. Warfarin 17-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 1581537-0 1992 Hydroxylation of warfarin by human cDNA-expressed cytochrome P-450: a role for P-4502C9 in the etiology of (S)-warfarin-drug interactions. Warfarin 111-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 1581537-3 1992 In order to identify the specific form(s) of human liver cytochrome P-450 involved in this particular toxicity, we have determined the metabolic profiles of 11 human cytochrome P-450 forms expressed in HepG2 cells toward both (R)- and (S)-warfarin. Warfarin 239-247 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 1581524-1 1992 The mechanism for cytochrome P-450 dependent conversion of hexahalogenated benzenes was investigated. Benzene 75-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-34 1581524-7 1992 On the basis of the results obtained, three of the four mechanisms proposed in the literature for the mechanism of the cytochrome P-450 dependent conversion of hexahalogenated benzenes to pentahalogenated phenols could be eliminated. Benzene 176-184 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 1581524-7 1992 On the basis of the results obtained, three of the four mechanisms proposed in the literature for the mechanism of the cytochrome P-450 dependent conversion of hexahalogenated benzenes to pentahalogenated phenols could be eliminated. Phenols 205-212 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 1581524-9 1992 On the basis of the results obtained a new hypothesis for the mechanism of cytochrome P-450 dependent conversion of hexahalogenated benzenes to pentahalogenated phenols was proposed. Benzene 132-140 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 1581524-9 1992 On the basis of the results obtained a new hypothesis for the mechanism of cytochrome P-450 dependent conversion of hexahalogenated benzenes to pentahalogenated phenols was proposed. Phenols 161-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 1571442-4 1992 Certain isozymes of the cytochrome P-450 system are selectively inhibited by some fluoroquinolones. Fluoroquinolones 82-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 1346997-0 1992 Characterization of the cytochrome P-450 gene family responsible for the N-dealkylation of the ergot alkaloid CQA 206-291 in humans. Nitrogen 73-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 1306804-0 1992 Cytochrome P450-dependent metabolism of dextromethorphan: fetal and adult studies. Dextromethorphan 40-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1346997-7 1992 This was substantiated by the finding that CQA metabolism was completely inhibited by a polyclonal antibody directed against a pregnenolone 16 alpha-carbonitrile-inducible cytochrome P-450 of rat liver. Amodiaquine 43-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 1346997-7 1992 This was substantiated by the finding that CQA metabolism was completely inhibited by a polyclonal antibody directed against a pregnenolone 16 alpha-carbonitrile-inducible cytochrome P-450 of rat liver. Pregnenolone 127-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 1346997-7 1992 This was substantiated by the finding that CQA metabolism was completely inhibited by a polyclonal antibody directed against a pregnenolone 16 alpha-carbonitrile-inducible cytochrome P-450 of rat liver. alpha-carbonitrile 143-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 1459043-0 1992 Relationship between activities of cytochrome P-450 monooxygenases in human placental microsomes and binding of benzo(a)pyrene metabolites to calf thymus DNA. benzo(a) 112-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 1459043-0 1992 Relationship between activities of cytochrome P-450 monooxygenases in human placental microsomes and binding of benzo(a)pyrene metabolites to calf thymus DNA. pyrene 120-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-51 1721276-0 1991 Effects of FK 506 on human hepatic microsomal cytochrome P-450-dependent drug metabolism in vitro. Tacrolimus 11-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 1596582-2 1992 Procarbazine is converted initially to azoprocarbazine, which is then N-oxidized through a cytochrome P-450-mediated process to a mixture of the positional isomers, benzylazoxyprocarbazine and methylazoxyprocarbazine. Procarbazine 0-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 1596582-2 1992 Procarbazine is converted initially to azoprocarbazine, which is then N-oxidized through a cytochrome P-450-mediated process to a mixture of the positional isomers, benzylazoxyprocarbazine and methylazoxyprocarbazine. azoprocarbazine 39-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 1596582-2 1992 Procarbazine is converted initially to azoprocarbazine, which is then N-oxidized through a cytochrome P-450-mediated process to a mixture of the positional isomers, benzylazoxyprocarbazine and methylazoxyprocarbazine. benzylazoxyprocarbazine 165-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 1596582-2 1992 Procarbazine is converted initially to azoprocarbazine, which is then N-oxidized through a cytochrome P-450-mediated process to a mixture of the positional isomers, benzylazoxyprocarbazine and methylazoxyprocarbazine. methylazoxyprocarbazine 193-216 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 1412450-0 1992 Roles of renal cytochrome P450-dependent arachidonic acid metabolites in hypertension. Arachidonic Acid 41-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-30 1412450-1 1992 Cytochrome P450 represents the third metabolic pathway of arachidonic acid giving rise to several biologically active compounds, such as 19-HETE, 20-HETE and EETs and their corresponding DHETs. Arachidonic Acid 58-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1412450-1 1992 Cytochrome P450 represents the third metabolic pathway of arachidonic acid giving rise to several biologically active compounds, such as 19-HETE, 20-HETE and EETs and their corresponding DHETs. 19-hydroxy-5,8,11,14-eicosatetraenoic acid 137-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1412450-1 1992 Cytochrome P450 represents the third metabolic pathway of arachidonic acid giving rise to several biologically active compounds, such as 19-HETE, 20-HETE and EETs and their corresponding DHETs. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 146-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1412450-1 1992 Cytochrome P450 represents the third metabolic pathway of arachidonic acid giving rise to several biologically active compounds, such as 19-HETE, 20-HETE and EETs and their corresponding DHETs. dhets 187-192 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 1955133-1 1991 Cimetidine binds to cytochrome P-450 and inhibits hepatic metabolism of various drugs in humans. Cimetidine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 20-36 1955133-6 1991 Concordantly, the calculated binding constant for the binding of cimetidine to a purified cytochrome P-450 with high tolbutamide hydroxylase activity was 4.4 mmol/L, whereas the calculated binding concentration constant for a purified cytochrome P-450-metabolizing nifedipine was 0.7 mmol/L. Cimetidine 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 1955133-6 1991 Concordantly, the calculated binding constant for the binding of cimetidine to a purified cytochrome P-450 with high tolbutamide hydroxylase activity was 4.4 mmol/L, whereas the calculated binding concentration constant for a purified cytochrome P-450-metabolizing nifedipine was 0.7 mmol/L. Cimetidine 65-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 235-251 1955133-6 1991 Concordantly, the calculated binding constant for the binding of cimetidine to a purified cytochrome P-450 with high tolbutamide hydroxylase activity was 4.4 mmol/L, whereas the calculated binding concentration constant for a purified cytochrome P-450-metabolizing nifedipine was 0.7 mmol/L. Nifedipine 265-275 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 90-106 1592216-10 1992 A significant bioactivation with a possibility of only limited detoxication via cytochrome P-450-dependent oxidation suggests that human fetus may be at greater risk from 1,2-dibromoethane toxicity than adult. Ethylene Dibromide 171-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-96 1594531-0 1992 Suicidal inactivation of human cytochrome P-450 by carbon tetrachloride and halothane in vitro. Carbon Tetrachloride 51-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 1594531-0 1992 Suicidal inactivation of human cytochrome P-450 by carbon tetrachloride and halothane in vitro. Halothane 76-85 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 1594531-1 1992 A significant loss of human cytochrome P-450 was observed during the anaerobic incubation of NADPH-reduced human liver microsomes obtained from surgical samples, in presence of carbon tetrachloride or halothane. NADP 93-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 1594531-1 1992 A significant loss of human cytochrome P-450 was observed during the anaerobic incubation of NADPH-reduced human liver microsomes obtained from surgical samples, in presence of carbon tetrachloride or halothane. Carbon Tetrachloride 177-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 1594531-1 1992 A significant loss of human cytochrome P-450 was observed during the anaerobic incubation of NADPH-reduced human liver microsomes obtained from surgical samples, in presence of carbon tetrachloride or halothane. Halothane 201-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 1594531-5 1992 The results indicate that human liver cytochrome P-450 can be inactivated reductively in vitro by CCl4 and halothane reactive metabolites and suggest that a suicide type of mechanism, similar to that which was recently demonstrated to occur, for both substrates, with rat liver microsomes (Manno et al. Halothane 107-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-54 1764041-6 1991 Cytochrome P-450 inhibitors prevented the thrombin-induced Mn2+ influx, with little effect on the Ca2+ mobilization from the intracellular stores. Manganese(2+) 59-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 1764041-10 1991 The activation of the plasma-membrane Ca2+ (Mn2+) pathway may take place by a mechanism involving microsomal cytochrome P-450, similar to that described previously in thymocytes [Alvarez, Montero & Garcia-Sancho (1991) Biochem. Manganese(2+) 44-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-125 1748033-0 1991 Clinical implications of drug interactions with the cytochrome P-450 enzyme system associated with omeprazole. Omeprazole 99-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 1748033-3 1991 Omeprazole contains a benzimidazole moiety and thus has the potential to interact with the cytochrome P-450 enzyme group. Omeprazole 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 1748033-3 1991 Omeprazole contains a benzimidazole moiety and thus has the potential to interact with the cytochrome P-450 enzyme group. benzimidazole 22-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 91-107 1748033-6 1991 Overall, the studies reviewed suggest that omeprazole has a differential affinity toward specific cytochrome P-450 isozymes. Omeprazole 43-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 1770192-4 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving cytochrome P-450: the newly discovered ethanol-inducible cytochrome P-450 (P-450IIE1) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 28-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 1770192-4 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving cytochrome P-450: the newly discovered ethanol-inducible cytochrome P-450 (P-450IIE1) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 28-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-183 1770192-4 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving cytochrome P-450: the newly discovered ethanol-inducible cytochrome P-450 (P-450IIE1) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 1770192-4 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving cytochrome P-450: the newly discovered ethanol-inducible cytochrome P-450 (P-450IIE1) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-183 1770192-4 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving cytochrome P-450: the newly discovered ethanol-inducible cytochrome P-450 (P-450IIE1) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 1770192-4 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving cytochrome P-450: the newly discovered ethanol-inducible cytochrome P-450 (P-450IIE1) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-183 1770192-4 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving cytochrome P-450: the newly discovered ethanol-inducible cytochrome P-450 (P-450IIE1) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-126 1770192-4 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving cytochrome P-450: the newly discovered ethanol-inducible cytochrome P-450 (P-450IIE1) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 167-183 1771946-1 1991 A cocktail of four model substances orally administered to humans is used for simultaneous characterization of phenobarbital and 3-methylcholanthrene inducible forms of cytochrome-P450 and two genetic polymorphisms, the debrisoquine hydroxylation and N-acetylation. Methylcholanthrene 129-149 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-184 1812615-2 1991 Benzidine and its derivatives were shown to induce those forms of cytochrome P-450 which were involved in accelerated oxidation of the carcinogenic drugs studied as well as affected the glutathione transferase, NADPH-dependent glutathione reductase activities and the activity of antioxidant system enzymes. benzidine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 1800391-1 1991 Urinary 6 beta-hydroxycortisol (6 beta OHF) is a specific marker of the induction of the cytochrome P-450 IIIA. 6 beta-hydroxycortisol 8-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 1800391-1 1991 Urinary 6 beta-hydroxycortisol (6 beta OHF) is a specific marker of the induction of the cytochrome P-450 IIIA. beta ohf 34-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-105 1811554-2 1991 The hypothesis that the mechanism of action of gemfibrozil involved the cytochrome P-450 system was tested by using ketoconazole, a P-450 inhibitor, which blocks the formation of endogenous polar sterols. Gemfibrozil 47-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 1811554-2 1991 The hypothesis that the mechanism of action of gemfibrozil involved the cytochrome P-450 system was tested by using ketoconazole, a P-450 inhibitor, which blocks the formation of endogenous polar sterols. Ketoconazole 116-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 1914510-0 1991 Immunocytochemical study of phenobarbital- and 3-methylcholanthrene-inducible cytochrome P450 isozymes in primary cultures of porcine ciliary epithelium. Phenobarbital 28-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 1951574-7 1991 Most of the imidazole and triazole derivatives are highly potent and selective inhibitors of the cytochrome P-450-dependent 14 alpha-demethylation of lanosterol (P-45014DM). imidazole 12-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 1951574-7 1991 Most of the imidazole and triazole derivatives are highly potent and selective inhibitors of the cytochrome P-450-dependent 14 alpha-demethylation of lanosterol (P-45014DM). Triazoles 26-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 1951574-7 1991 Most of the imidazole and triazole derivatives are highly potent and selective inhibitors of the cytochrome P-450-dependent 14 alpha-demethylation of lanosterol (P-45014DM). Lanosterol 150-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 1951574-9 1991 The triazole antifungals, terconazole and itraconazole, combine a high affinity for Candida P-45014DM with an exceptionally low effect on mammalian cytochrome P-450. Triazoles 4-12 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 1951574-9 1991 The triazole antifungals, terconazole and itraconazole, combine a high affinity for Candida P-45014DM with an exceptionally low effect on mammalian cytochrome P-450. terconazole 26-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 1951574-9 1991 The triazole antifungals, terconazole and itraconazole, combine a high affinity for Candida P-45014DM with an exceptionally low effect on mammalian cytochrome P-450. Itraconazole 42-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 1930182-3 1991 In cases where iminium-enamine coupling is sterically prevented, the iminium in equilibrium with enamine species can be studied independently and are found to be more potent metabolism-dependent inactivators of cytochrome P-450 than are the corresponding parent amines. iminium-enamine 15-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-227 1930182-3 1991 In cases where iminium-enamine coupling is sterically prevented, the iminium in equilibrium with enamine species can be studied independently and are found to be more potent metabolism-dependent inactivators of cytochrome P-450 than are the corresponding parent amines. iminium 15-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-227 1930182-3 1991 In cases where iminium-enamine coupling is sterically prevented, the iminium in equilibrium with enamine species can be studied independently and are found to be more potent metabolism-dependent inactivators of cytochrome P-450 than are the corresponding parent amines. cyclopentyl enamine 23-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-227 1930182-3 1991 In cases where iminium-enamine coupling is sterically prevented, the iminium in equilibrium with enamine species can be studied independently and are found to be more potent metabolism-dependent inactivators of cytochrome P-450 than are the corresponding parent amines. Amines 262-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-227 1686246-4 1991 The activities were NADPH dependent and inhibited by 1-aminobenzotriazole, a potent cytochrome P-450 suicide substrate inhibitor. NADP 20-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 1686246-4 1991 The activities were NADPH dependent and inhibited by 1-aminobenzotriazole, a potent cytochrome P-450 suicide substrate inhibitor. 1-aminobenzotriazole 53-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 1876834-6 1991 The results establish a new catalytic activity for a cytochrome P-450 and illustrate the cooperation of different oxygenases in pathways of fatty acid metabolism. Fatty Acids 140-150 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 1752284-0 1991 Cyclosporin-A-induced lipid peroxidation in human liver microsomes and its influence on cytochrome P-450. Cyclosporine 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 1752284-1 1991 The present in vitro study using human liver tissue was performed to investigate the effect of cyclosporin A on lipid peroxidation and cytochrome P-450 concentration in isolated liver microsomes. Cyclosporine 95-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 135-151 1752284-9 1991 Furthermore, cyclosporin-A-induced microsomal lipid peroxidation was accompanied by a significant dose-dependent decline of the microsomal cytochrome P-450 content. Cyclosporine 13-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-155 1752284-10 1991 At a cyclosporin A concentration of 300 micrograms ml-1, cytochrome P-450 content was decreased to 49% in comparison to control values. Cyclosporine 5-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 57-73 1752284-11 1991 In the presence of reduced glutathione, cyclosporin A decreased the cytochrome P-450 concentration only to 79% (P less than 0.05). Glutathione 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 1752284-11 1991 In the presence of reduced glutathione, cyclosporin A decreased the cytochrome P-450 concentration only to 79% (P less than 0.05). Cyclosporine 40-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 1650691-3 1991 Most investigations on this topic have focused on two aspects: ethanol"s capacity to induce the cytochrome P-450-dependent microsomal biotransformation system and its interference with at least one DNA repair mechanism. Ethanol 63-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 96-112 1914510-0 1991 Immunocytochemical study of phenobarbital- and 3-methylcholanthrene-inducible cytochrome P450 isozymes in primary cultures of porcine ciliary epithelium. Methylcholanthrene 47-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 1771239-1 1991 Epoxides and fatty acid diols derived from arachidonate by the action of cytochrome P-450 appear in human urine and have biological activities. Epoxy Compounds 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 1720447-2 1991 Multiple isoforms of cytochrome P450 (P450) have been discovered that are important biomedically to drug pharmacokinetics, chemical carcinogenesis, and metabolism of endogenous agents such as steroids, arachidonic acids, and prostaglandins. Steroids 192-200 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 1720447-2 1991 Multiple isoforms of cytochrome P450 (P450) have been discovered that are important biomedically to drug pharmacokinetics, chemical carcinogenesis, and metabolism of endogenous agents such as steroids, arachidonic acids, and prostaglandins. Arachidonic Acids 202-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 1720447-2 1991 Multiple isoforms of cytochrome P450 (P450) have been discovered that are important biomedically to drug pharmacokinetics, chemical carcinogenesis, and metabolism of endogenous agents such as steroids, arachidonic acids, and prostaglandins. Prostaglandins 225-239 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 1720069-6 1991 The free radical scavenger, alpha-tocopherol, and the xanthine oxidase inhibitor, allopurinol, also prevented the loss of cytochrome P-450 mediated by the interferon inducer poly rI.rC. alpha-Tocopherol 28-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 1720069-6 1991 The free radical scavenger, alpha-tocopherol, and the xanthine oxidase inhibitor, allopurinol, also prevented the loss of cytochrome P-450 mediated by the interferon inducer poly rI.rC. Allopurinol 82-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 122-138 1680663-0 1991 Blood levels of propylene oxide during propylene inhalation and effect on hepatic and nasal cytochrome P-450 concentrations. propylene oxide 16-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 1680663-0 1991 Blood levels of propylene oxide during propylene inhalation and effect on hepatic and nasal cytochrome P-450 concentrations. propylene 16-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 92-108 1857342-1 1991 Purification and immunoinhibition studies have suggested that the hydroxylations of (S)-mephenytoin and tolbutamide are catalyzed by rather similar forms of human liver cytochrome P-450 (P-450). Mephenytoin 84-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 1857342-1 1991 Purification and immunoinhibition studies have suggested that the hydroxylations of (S)-mephenytoin and tolbutamide are catalyzed by rather similar forms of human liver cytochrome P-450 (P-450). Tolbutamide 104-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 169-185 1650250-3 1991 Thus, chloroperoxidase shares many spectroscopic properties with cytochrome P-450, the only other known thiolate-ligated heme protein. thiolate 104-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 1650250-3 1991 Thus, chloroperoxidase shares many spectroscopic properties with cytochrome P-450, the only other known thiolate-ligated heme protein. Heme 121-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 65-81 1906712-4 1991 The calcium-stores-regulated Mn2+ influx, including that induced by agonists, was prevented by cytochrome P-450 inhibitors. Calcium 4-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 1906712-4 1991 The calcium-stores-regulated Mn2+ influx, including that induced by agonists, was prevented by cytochrome P-450 inhibitors. Manganese(2+) 29-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 1906712-5 1991 We propose that agonist-induced Ca2+ (Mn2+) influx in human neutrophils is secondary to the emptying of the intracellular stores which, in turn, activates plasma-membrane Ca2+ channels by a mechanism involving microsomal cytochrome P-450, similar to that described previously in thymocytes [Alvarez, Montero & Garcia-Sancho (1991) Biochem. Manganese(2+) 38-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 221-237 1771239-1 1991 Epoxides and fatty acid diols derived from arachidonate by the action of cytochrome P-450 appear in human urine and have biological activities. fatty acid diols 13-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 1771239-1 1991 Epoxides and fatty acid diols derived from arachidonate by the action of cytochrome P-450 appear in human urine and have biological activities. Arachidonic Acid 43-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 1771239-3 1991 We investigated whether dietary eicosapentaenoic acid could also be metabolized by cytochrome P-450, by assessing the excretion of its vicinal diols. Eicosapentaenoic Acid 32-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 1771239-3 1991 We investigated whether dietary eicosapentaenoic acid could also be metabolized by cytochrome P-450, by assessing the excretion of its vicinal diols. vicinal diols 135-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 83-99 1949908-15 1991 Moreover, both these pathways involve the cytochrome P-450 system and can be induced with phenobarbital. Phenobarbital 90-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-58 2040591-10 1991 Thus aldosterone synthase cytochrome P-450, a distinct species from cytochrome P-450(11 beta), is responsible for the biosynthesis of aldosterone in the human, at least in patients suffering from primary aldosteronism. Aldosterone 5-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-42 2040591-10 1991 Thus aldosterone synthase cytochrome P-450, a distinct species from cytochrome P-450(11 beta), is responsible for the biosynthesis of aldosterone in the human, at least in patients suffering from primary aldosteronism. Aldosterone 5-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-84 2029514-0 1991 CO binding studies of engineered cytochrome P-450ds: effects of mutations at putative distal sites in the presence of polycyclic hydrocarbons. Hydrocarbons, Cyclic 118-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 2004631-0 1991 Carbamazepine dose requirements during stiripentol therapy: influence of cytochrome P-450 inhibition by stiripentol. stiripentol 104-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 2061924-1 1991 A series of 21 different 4-substituted 2,6-dimethyl-3-(alkoxycarbonyl)-1,4-dihydropyridines was considered with regard to oxidation to pyridine derivatives by human liver microsomal cytochrome P-450 (P-450). 4-substituted 2,6-dimethyl-3-(alkoxycarbonyl)-1,4-dihydropyridines 25-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-198 2061924-1 1991 A series of 21 different 4-substituted 2,6-dimethyl-3-(alkoxycarbonyl)-1,4-dihydropyridines was considered with regard to oxidation to pyridine derivatives by human liver microsomal cytochrome P-450 (P-450). pyridine 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 182-198 2049228-14 1991 These in vitro studies are consistent with phenytoin 4-hydroxylation and tolbutamide methylhydroxylation being catalysed by the same cytochrome P450 isozyme(s) in human liver microsomes. Phenytoin 43-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 2066864-3 1991 The cytochrome P-450 preparation had an apparent molecular weight of 51,500 as judged by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Sodium Dodecyl Sulfate 89-111 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2066864-3 1991 The cytochrome P-450 preparation had an apparent molecular weight of 51,500 as judged by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. polyacrylamide 112-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2049228-14 1991 These in vitro studies are consistent with phenytoin 4-hydroxylation and tolbutamide methylhydroxylation being catalysed by the same cytochrome P450 isozyme(s) in human liver microsomes. Tolbutamide 73-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-148 1846080-4 1991 A good fit of the absorption spectra of six different cytochrome P-450 proteins in the presence and absence of substrates was found, indicating a similar pi-electron structure of the porphyrin and a similar chemical nature of the nearest coordination sphere of the iron in all cytochrome P-450 proteins. Iron 265-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 1846317-4 1991 H2O2 production was inhibited by diphenyleneiodonium, a flavoprotein binder (concentration producing 50% inhibition, 0.3 microM), and diethyldithiocarbamate, a divalent cation chelator (concentration producing 50% inhibition, 3 microM), but not by cyanide or azide, inhibitors of electron transport, or by agents that inhibit xanthine oxidase, polyamine oxidase, or cytochrome P450. diphenyleneiodonium 33-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 366-381 1846317-4 1991 H2O2 production was inhibited by diphenyleneiodonium, a flavoprotein binder (concentration producing 50% inhibition, 0.3 microM), and diethyldithiocarbamate, a divalent cation chelator (concentration producing 50% inhibition, 3 microM), but not by cyanide or azide, inhibitors of electron transport, or by agents that inhibit xanthine oxidase, polyamine oxidase, or cytochrome P450. Ditiocarb 134-156 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 366-381 2004043-2 1991 All the progestogens are acetylenic steroids and previous studies have indicated the potential of acetylenic steroids to cause mechanism-based or "suicide" inactivation of cytochrome P-450. acetylenic steroids 25-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 2004043-2 1991 All the progestogens are acetylenic steroids and previous studies have indicated the potential of acetylenic steroids to cause mechanism-based or "suicide" inactivation of cytochrome P-450. acetylenic steroids 98-117 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 1846317-4 1991 H2O2 production was inhibited by diphenyleneiodonium, a flavoprotein binder (concentration producing 50% inhibition, 0.3 microM), and diethyldithiocarbamate, a divalent cation chelator (concentration producing 50% inhibition, 3 microM), but not by cyanide or azide, inhibitors of electron transport, or by agents that inhibit xanthine oxidase, polyamine oxidase, or cytochrome P450. Hydrogen Peroxide 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 366-381 1920538-2 1991 Cytochrome P-450 catalyzes oxidations of a wide variety of compounds including steroids, drugs and toxicants. Steroids 79-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 1846080-4 1991 A good fit of the absorption spectra of six different cytochrome P-450 proteins in the presence and absence of substrates was found, indicating a similar pi-electron structure of the porphyrin and a similar chemical nature of the nearest coordination sphere of the iron in all cytochrome P-450 proteins. Iron 265-269 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 277-293 1669007-5 1991 These reactions are elevated after chronic ethanol consumption, due in part, to induction of a unique isozyme of cytochrome P-450. Ethanol 43-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-129 1819909-5 1991 The phenobarbital-induced BND activity and the cytochrome P-450 content were not changed, while the beta-naphthoflavone-induced EROD activity and cytochrome P-450 and cytochrome b5 content decreased, the ECOD activity remaining unchanged. beta-Naphthoflavone 100-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 146-180 1819909-6 1991 Nifedipine administered for three days significantly increased the BND activity and the cytochrome P-450 content. Nifedipine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 88-104 1819909-7 1991 The three-day administration of nifedipine plus phenobarbital resulted in a further increase in the BND activity and the cytochrome P-450 content. Nifedipine 32-42 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 1819909-7 1991 The three-day administration of nifedipine plus phenobarbital resulted in a further increase in the BND activity and the cytochrome P-450 content. Phenobarbital 48-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 1819909-9 1991 Nifedipine administered at a single dose, one hour before beta-naphthoflavone had no effect per se but potentiated the enzyme-inducing effect of beta-naphthoflavone on the EROD activity and the cytochrome P-450 content but not on the ECOD activity. Nifedipine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-210 1819909-9 1991 Nifedipine administered at a single dose, one hour before beta-naphthoflavone had no effect per se but potentiated the enzyme-inducing effect of beta-naphthoflavone on the EROD activity and the cytochrome P-450 content but not on the ECOD activity. beta-Naphthoflavone 145-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 194-210 1805731-0 1991 Influence of inducers of cytochrome P-450 on dealkylation of 7-alkoxycoumarins. 7-alkoxycoumarins 61-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 1709343-2 1991 This loss in drug metabolism is due to the loss of the cytochrome P-450 component of the mixed function oxidase (the enzyme system primarily responsible for the oxidation of drugs, carcinogens and certain classes of endogenous substances such as steroids, fatty acids and prostaglandins). Steroids 246-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 1709343-2 1991 This loss in drug metabolism is due to the loss of the cytochrome P-450 component of the mixed function oxidase (the enzyme system primarily responsible for the oxidation of drugs, carcinogens and certain classes of endogenous substances such as steroids, fatty acids and prostaglandins). Fatty Acids 256-267 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 1709343-2 1991 This loss in drug metabolism is due to the loss of the cytochrome P-450 component of the mixed function oxidase (the enzyme system primarily responsible for the oxidation of drugs, carcinogens and certain classes of endogenous substances such as steroids, fatty acids and prostaglandins). Prostaglandins 272-286 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 2014968-7 1991 However, careful clinical monitoring is necessary with all patients, irrespective of age, taking ciprofloxacin concomitantly with drugs primarily eliminated by the cytochrome P-450 system. Ciprofloxacin 97-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-180 1905908-0 1991 Cytochrome P-450 metabolism of arachidonic acid. Arachidonic Acid 31-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2009574-4 1991 Cytochrome P-450 dependent monooxygenation at a non-fluorinated para position in (fluoro)aniline derivatives proceeds by formation of the para hydroxylated derivative as the primary metabolite. (fluoro)aniline 81-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2009574-6 1991 Thus, for fluoroanilines with a fluorine substituent at the para position bioactivation to the reactive benzoquinoneimine can be a direct result of the cytochrome P-450 dependent conversion. fluoroanilines 10-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-168 2009574-6 1991 Thus, for fluoroanilines with a fluorine substituent at the para position bioactivation to the reactive benzoquinoneimine can be a direct result of the cytochrome P-450 dependent conversion. Fluorine 32-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-168 2009574-6 1991 Thus, for fluoroanilines with a fluorine substituent at the para position bioactivation to the reactive benzoquinoneimine can be a direct result of the cytochrome P-450 dependent conversion. benzoquinoneimine 104-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 152-168 16840049-2 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450; this newly discovered ethanol-inducible cytochrome P-450 (P-450 IIEi) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 28-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 1685398-10 1991 It is concluded that treatment with a commercial PCB mixture resulted in the induction of several isoforms of pigeon hepatic cytochrome P-450 in a fashion that is likely to be similar to that reported for mammals. Polychlorinated Biphenyls 49-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 125-141 16840049-2 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450; this newly discovered ethanol-inducible cytochrome P-450 (P-450 IIEi) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 16840049-2 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450; this newly discovered ethanol-inducible cytochrome P-450 (P-450 IIEi) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-195 2043050-0 1991 Effects of acetone administration on cytochrome P-450-dependent monooxygenases in hamster liver, kidney, and lung. Acetone 11-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 2043050-3 1991 Administration of acetone caused significant increases of cytochrome P-450 and cytochrome b5 contents in liver microsomes. Acetone 18-25 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 2043050-9 1991 Gel electrophoresis of liver and kidney microsomes from control and acetone-treated hamsters revealed that acetone treatment enhanced the intensity of a protein band(s) in the cytochrome P-450 molecular weight region. Acetone 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-192 16840049-2 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450; this newly discovered ethanol-inducible cytochrome P-450 (P-450 IIEi) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 2043050-9 1991 Gel electrophoresis of liver and kidney microsomes from control and acetone-treated hamsters revealed that acetone treatment enhanced the intensity of a protein band(s) in the cytochrome P-450 molecular weight region. Acetone 107-114 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 176-192 16840049-2 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450; this newly discovered ethanol-inducible cytochrome P-450 (P-450 IIEi) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-195 16840049-2 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450; this newly discovered ethanol-inducible cytochrome P-450 (P-450 IIEi) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 28-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-195 2043050-10 1991 Immunoblotting of the microsomal proteins showed that the protein band induced by acetone in hamster liver, kidney and lung was cross-reactive with antibody raised against ethanol-inducible human liver cytochrome P-450. Acetone 82-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 2043050-10 1991 Immunoblotting of the microsomal proteins showed that the protein band induced by acetone in hamster liver, kidney and lung was cross-reactive with antibody raised against ethanol-inducible human liver cytochrome P-450. Ethanol 172-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 202-218 16840049-2 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450; this newly discovered ethanol-inducible cytochrome P-450 (P-450 IIEi) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-137 2043050-11 1991 These results demonstrate that acetone has the ability to uniformly induce a specific form of cytochrome P-450, designated as IIE1, and to cause differential changes of monooxygenase activities in the hamster tissues. Acetone 31-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 16840049-2 1991 It was also determined that ethanol can be oxidized by a microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450; this newly discovered ethanol-inducible cytochrome P-450 (P-450 IIEi) contributes to ethanol metabolism, tolerance, energy wastage (with associated weight loss), and the selective hepatic perivenular toxicity of various xenobiotics. Ethanol 68-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 179-195 1937350-11 1991 The cytochrome P-450 isozyme responsible for debrisoquine hydroxylation is of high affinity-low capacity character, which means that it can be saturated under certain circumstances. Debrisoquin 45-57 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 1864266-13 1991 The possibility that myrcene exerts its antimutagenic activity by inhibiting certain forms of the cytochrome P-450 isoenzymes required for activation of premutagens and precarcinogenes is discussed. myrcene 21-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 1667771-0 1991 Cytochrome P-450- and peroxidase-dependent activation of procarbazine and iproniazid in mammalian cells. Procarbazine 57-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-32 1667771-0 1991 Cytochrome P-450- and peroxidase-dependent activation of procarbazine and iproniazid in mammalian cells. Iproniazid 74-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-32 1792338-8 1991 PB and TAO increased cytochrome P-450 and associated enzyme activities, in particular those related to cytochrome P-450p or P-450NF (including oestradiol 2-hydroxylation) in the human liver. Estradiol 143-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 1667771-3 1991 Both peroxidases and cytochrome P-450 have been reported to catalyze biotransformation of hydrazines. Hydrazines 90-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 1667771-5 1991 However, in HL60 cells, procarbazine was metabolized by myeloperoxidase while iproniazid was metabolized mostly by the cytochrome P-450 system. Iproniazid 78-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 119-135 1761357-2 1991 We now report that IFN-alpha-Con1, which was constructed from the most frequently observed amino acid sequences in human alpha-interferon subtypes, causes a loss in cytochrome P-450 which could be prevented by pretreating animals with either puromycin or actinomycin D. Puromycin 242-251 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-181 1761357-2 1991 We now report that IFN-alpha-Con1, which was constructed from the most frequently observed amino acid sequences in human alpha-interferon subtypes, causes a loss in cytochrome P-450 which could be prevented by pretreating animals with either puromycin or actinomycin D. Dactinomycin 255-268 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 165-181 1812974-3 1991 The first hypothesis concerns the role of the oxygen binding hemoprotein cytochrome P-450. Oxygen 46-52 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-89 1812974-10 1991 The possibility exists that changes in both the cytochrome P-450 activity and in the rate of electrons flow in the respiratory chain may alter the amount of oxygen radicals in the cells and, similarly as in the "oxygen radicals" hypothesis, govern calcium channels through the control of the redox status of these channels. Reactive Oxygen Species 157-172 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 1812974-10 1991 The possibility exists that changes in both the cytochrome P-450 activity and in the rate of electrons flow in the respiratory chain may alter the amount of oxygen radicals in the cells and, similarly as in the "oxygen radicals" hypothesis, govern calcium channels through the control of the redox status of these channels. Reactive Oxygen Species 212-227 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 1852787-5 1991 These considerations are illustrated by examination of the generation and fate of enzyme- and substrate-derived free radicals at various stages in the catalytic cycles of two monooxygenases important in xenobiotic biotransformation, dopamine beta-hydroxylase and cytochrome P-450. Free Radicals 112-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 263-279 1965943-0 1990 The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces changes in the heme spin state of microsomal cytochrome P-450. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 15-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 1808966-1 1991 Caffeine is mainly metabolized by 3-methylcholanthreneinducible cytochrome P-450, whereas metamizol (Analgin) is probably mainly metabolized by the phenobarbital inducible cytochrome P-450 family. Caffeine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-80 1808966-1 1991 Caffeine is mainly metabolized by 3-methylcholanthreneinducible cytochrome P-450, whereas metamizol (Analgin) is probably mainly metabolized by the phenobarbital inducible cytochrome P-450 family. Dipyrone 90-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 1808966-1 1991 Caffeine is mainly metabolized by 3-methylcholanthreneinducible cytochrome P-450, whereas metamizol (Analgin) is probably mainly metabolized by the phenobarbital inducible cytochrome P-450 family. Phenobarbital 148-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-188 1808966-2 1991 Therefore the elimination of caffeine from serum and the elimination of the main metabolites of metamizol in urine reflect the activity of these two cytochrome P-450 families. Caffeine 29-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 1808966-2 1991 Therefore the elimination of caffeine from serum and the elimination of the main metabolites of metamizol in urine reflect the activity of these two cytochrome P-450 families. Dipyrone 96-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 149-165 2269307-2 1990 Cytochrome P-450 (P450) NF, a member of the P450 IIIA subfamily, is the major contributor to the oxidation of the calcium-channel blocker nifedipine in human liver microsomes. Nifedipine 138-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-26 1965943-4 1990 EPR studies confirmed that MPTP is a type II substrate for the forms of cytochrome P-450 with which it interacts and causes a shift from the high spin state of cytochrome P-450 to the low spin state. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 1965943-4 1990 EPR studies confirmed that MPTP is a type II substrate for the forms of cytochrome P-450 with which it interacts and causes a shift from the high spin state of cytochrome P-450 to the low spin state. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 27-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 160-176 1965943-5 1990 MPTP is, thus, likely to be an effective inhibitor of cytochrome P-450. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 1768430-7 1991 The Ah-receptor binding compounds, such as coal tar constituents, or 3-methylcholanthrene induce cytochrome P-450-dependent activities such as aryl hydrocarbon hydroxylase or 7-ethoxyresorufin-O-de-ethylase and NQR, whereas butyl hydroxytoluol, which does not bind to the Ah receptor, induces only NQR. Methylcholanthrene 69-89 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 1768430-7 1991 The Ah-receptor binding compounds, such as coal tar constituents, or 3-methylcholanthrene induce cytochrome P-450-dependent activities such as aryl hydrocarbon hydroxylase or 7-ethoxyresorufin-O-de-ethylase and NQR, whereas butyl hydroxytoluol, which does not bind to the Ah receptor, induces only NQR. butyl hydroxytoluol 224-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 1965943-0 1990 The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces changes in the heme spin state of microsomal cytochrome P-450. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 61-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 1965943-0 1990 The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces changes in the heme spin state of microsomal cytochrome P-450. Heme 90-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 120-136 1965943-1 1990 In vitro studies on the nature of interaction of the neurotoxin MPTP with hepatic microsomal cytochrome P-450 were carried out. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 64-68 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 1965943-3 1990 Scatchard analysis of MPTP-cytochrome P-450 binding suggested that MPTP binds to at least 2 species of cytochrome P-450--a high affinity binding species with an apparent spectral dissociation constant (Ks) of 372 microM and a low affinity species with Ks of 37.6 mM. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 22-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-43 1965943-3 1990 Scatchard analysis of MPTP-cytochrome P-450 binding suggested that MPTP binds to at least 2 species of cytochrome P-450--a high affinity binding species with an apparent spectral dissociation constant (Ks) of 372 microM and a low affinity species with Ks of 37.6 mM. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 22-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 2099150-0 1990 Cytochrome P-450 catalyzed redox cycling of orthophenylphenol. 2-phenylphenol 44-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2099150-4 1990 Phenyl 2,5"-p-quinone was reduced to phenylhydroquinone by cytochrome P-450 reductase. phenyl 2,5"-p-quinone 0-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 2099150-4 1990 Phenyl 2,5"-p-quinone was reduced to phenylhydroquinone by cytochrome P-450 reductase. phenylhydroquinone 37-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 2099150-5 1990 This study provides direct evidence of cytochrome P-450 catalyzed redox cycling of o-phenylphenol. 2-phenylphenol 83-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 1965943-3 1990 Scatchard analysis of MPTP-cytochrome P-450 binding suggested that MPTP binds to at least 2 species of cytochrome P-450--a high affinity binding species with an apparent spectral dissociation constant (Ks) of 372 microM and a low affinity species with Ks of 37.6 mM. Potassium 202-204 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 1965943-3 1990 Scatchard analysis of MPTP-cytochrome P-450 binding suggested that MPTP binds to at least 2 species of cytochrome P-450--a high affinity binding species with an apparent spectral dissociation constant (Ks) of 372 microM and a low affinity species with Ks of 37.6 mM. Potassium 252-254 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-119 2268369-0 1990 Immunochemical detection of human liver cytochrome P450 forms related to phenobarbital-inducible forms in the mouse. Phenobarbital 73-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-55 2262908-0 1990 Lidocaine metabolism by human cytochrome P-450s purified from hepatic microsomes: comparison of those with rat hepatic cytochrome P-450s. Lidocaine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 2148293-0 1990 The effect of substrates and inhibitors of cytochrome P-450 on the NADPH inhibition of the ATP-dependent, hepatic, microsomal calcium pump. NADP 67-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2148293-0 1990 The effect of substrates and inhibitors of cytochrome P-450 on the NADPH inhibition of the ATP-dependent, hepatic, microsomal calcium pump. Adenosine Triphosphate 91-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2148293-1 1990 In hepatic microsomes one or more isozymes of cytochrome P-450 inhibits the ATP-dependent Ca2+ pump. Adenosine Triphosphate 76-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2172779-3 1990 We have previously shown that cytochrome P-450 in the liver metabolizes sulfamethoxazole to its hydroxylamine metabolite. Sulfamethoxazole 72-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 2256435-2 1990 Antipyrine is a useful marker drug for cytochrome P-450 dependent hepatic drug metabolism. Antipyrine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-55 1981505-0 1990 Quinolone inhibition of cytochrome P-450-dependent caffeine metabolism in human liver microsomes. Quinolones 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 1981505-0 1990 Quinolone inhibition of cytochrome P-450-dependent caffeine metabolism in human liver microsomes. Caffeine 51-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-40 1981505-7 1990 The results indicate that the reduction of caffeine clearance by concomitant quinolone application observed in vivo is primarily due to a competitive interaction of the inhibiting quinolones with the cytochrome P-450 isoenzyme(s) mediating caffeine demethylation. Caffeine 43-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-216 1981505-7 1990 The results indicate that the reduction of caffeine clearance by concomitant quinolone application observed in vivo is primarily due to a competitive interaction of the inhibiting quinolones with the cytochrome P-450 isoenzyme(s) mediating caffeine demethylation. Quinolones 77-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-216 1981505-7 1990 The results indicate that the reduction of caffeine clearance by concomitant quinolone application observed in vivo is primarily due to a competitive interaction of the inhibiting quinolones with the cytochrome P-450 isoenzyme(s) mediating caffeine demethylation. Quinolones 180-190 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-216 1981505-7 1990 The results indicate that the reduction of caffeine clearance by concomitant quinolone application observed in vivo is primarily due to a competitive interaction of the inhibiting quinolones with the cytochrome P-450 isoenzyme(s) mediating caffeine demethylation. Caffeine 240-248 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 200-216 1981539-8 1990 These results suggest that cytochrome P-450, rather than alcohol dehydrogenase, was responsible for the metabolism of benzyl alcohol to benzaldehyde. Benzyl Alcohol 118-132 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-78 1981539-8 1990 These results suggest that cytochrome P-450, rather than alcohol dehydrogenase, was responsible for the metabolism of benzyl alcohol to benzaldehyde. benzaldehyde 136-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-78 2172779-3 1990 We have previously shown that cytochrome P-450 in the liver metabolizes sulfamethoxazole to its hydroxylamine metabolite. Hydroxylamine 96-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 2122459-0 1990 Evidence for involvement of multiple forms of cytochrome P-450 in aflatoxin B1 metabolism in human liver. Aflatoxin B1 66-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2233694-0 1990 Stereoselective S-oxygenation of 2-aryl-1,3-dithiolanes by the flavin-containing and cytochrome P-450 monooxygenases. 2-aryl-1,3-dithiolanes 33-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 2226459-0 1990 Effect of the tyrosine 96 hydrogen bond on the inactivation of cytochrome P-450cam induced by hydrostatic pressure. Tyrosine 14-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 2226459-0 1990 Effect of the tyrosine 96 hydrogen bond on the inactivation of cytochrome P-450cam induced by hydrostatic pressure. Hydrogen 26-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 2233694-0 1990 Stereoselective S-oxygenation of 2-aryl-1,3-dithiolanes by the flavin-containing and cytochrome P-450 monooxygenases. 4,6-dinitro-o-cresol 63-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 2233694-5 1990 For S-oxide formation in microsomes, the data provided evidence for a minor role of cytochrome P-450 in S-oxide formation, but the flavin-containing monooxygenase was mainly responsible for production of S-oxide. Oxides 4-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 2242730-2 1990 Monensin was seen to cause a slight depression in the amount of cytochrome P-450 and cytochrome b5 as well as in the activities of aniline-p-hydroxylase, p-nitrophenol-hydroxylase and p-nitroanisole-O-demethylase. Monensin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-98 2261467-1 1990 Cytochrome P-450cam reacts with phenyldiazene (PhN = NH), or less efficiently with phenylhydrazine, to give a catalytically inactive complex with an absorption maximum at 474 nm. phenyldiazene 32-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2261467-1 1990 Cytochrome P-450cam reacts with phenyldiazene (PhN = NH), or less efficiently with phenylhydrazine, to give a catalytically inactive complex with an absorption maximum at 474 nm. phenylhydrazine 83-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2078633-1 1990 It has been found that 3-amino-7-dimethylamino-2-methylphenazine (neutral red, NR) is responsible for the electron transport from the cathode to adrenodoxin (Ad) and cytochrome P-450 (P-450scc) from adrenal cortex mitochondria inaccessible to direct electrochemical reduction under native conditions. Neutral Red Base 23-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 166-182 2260347-2 1990 Its mechanism of action, like that of other azoles, involves interruption of the conversion of lanosterol to ergosterol via binding to fungal cytochrome P-450 and subsequent disruption of fungal membranes. Azoles 44-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-158 2260347-2 1990 Its mechanism of action, like that of other azoles, involves interruption of the conversion of lanosterol to ergosterol via binding to fungal cytochrome P-450 and subsequent disruption of fungal membranes. Lanosterol 95-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-158 2260347-2 1990 Its mechanism of action, like that of other azoles, involves interruption of the conversion of lanosterol to ergosterol via binding to fungal cytochrome P-450 and subsequent disruption of fungal membranes. Ergosterol 109-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-158 1981700-0 1990 Expressing the specificity and the potency of cytochrome P-450 inducers toward 7-ethoxyresorufin. ethoxyresorufin 79-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2242730-3 1990 Tiamulin induced a moderate increase in the amount of cytochrome P-450 and in the activities of aniline-p-hydroxylase, p-nitrophenol-hydroxylase and aminopyrine-N-demethylase. tiamulin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-70 2242730-4 1990 The combined administration of monensin and tiamulin resulted in marked induction of the microsomal enzymes; the amount of cytochrome P-450 reduced by metyrapone or carbon monoxide increased 2.5 or 2-times, respectively, and the activities of the tested microsomal hydroxylases and demethylases showed also an expressed increase. Metyrapone 151-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 2242730-4 1990 The combined administration of monensin and tiamulin resulted in marked induction of the microsomal enzymes; the amount of cytochrome P-450 reduced by metyrapone or carbon monoxide increased 2.5 or 2-times, respectively, and the activities of the tested microsomal hydroxylases and demethylases showed also an expressed increase. Carbon Monoxide 165-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 123-139 2290428-2 1990 The benzene derivatives are typical substrates of cytochrome P-450 catalyzed aromatic oxidation. Benzene 4-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 2079689-5 1990 The major activation step is believed to be the oxygenation of the alpha-carbon catalysed by a Cytochrome P-450-dependent enzyme system commonly know as NDMA-demethylase. Carbon 73-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 2079689-6 1990 Studies on the enzymology of NDMA metabolism show that some Cytochrome P-450 isozymes exhibit significant NDMA-demethylase activity only at high NDMA concentrations. Dimethylnitrosamine 29-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 2079689-6 1990 Studies on the enzymology of NDMA metabolism show that some Cytochrome P-450 isozymes exhibit significant NDMA-demethylase activity only at high NDMA concentrations. Dimethylnitrosamine 106-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 2168169-1 1990 The rotation of cytochrome P-450 LM2 (CYPIIB4) incorporated into large microsomal-like lipid vesicles was investigated by saturation transfer EPR using 15N- and 2H-substituted spin labels. 15n 152-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 2219115-6 1990 3-Methylcholanthrene pretreatment increased and pretreatment with cobalt chloride and piperonyl butoxide decreased the hepatic covalent binding of 3HAA, indicating the involvement of cytochrome P450 in the formation of the 3HAA reactive metabolite. Methylcholanthrene 0-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-198 2219115-6 1990 3-Methylcholanthrene pretreatment increased and pretreatment with cobalt chloride and piperonyl butoxide decreased the hepatic covalent binding of 3HAA, indicating the involvement of cytochrome P450 in the formation of the 3HAA reactive metabolite. cobaltous chloride 66-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-198 2219115-6 1990 3-Methylcholanthrene pretreatment increased and pretreatment with cobalt chloride and piperonyl butoxide decreased the hepatic covalent binding of 3HAA, indicating the involvement of cytochrome P450 in the formation of the 3HAA reactive metabolite. Piperonyl Butoxide 86-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 183-198 2168169-1 1990 The rotation of cytochrome P-450 LM2 (CYPIIB4) incorporated into large microsomal-like lipid vesicles was investigated by saturation transfer EPR using 15N- and 2H-substituted spin labels. Deuterium 161-163 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 2214784-1 1990 An oral antimycotic agent, ketoconazole has been demonstrated to be an inhibitor of cytochrome P-450-dependent monooxygenases. Ketoconazole 27-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-100 2238708-6 1990 The evidence presented in this paper supports a different mechanism involving chronic induction of the microsomal cytochrome P-450 system, mobilization of hepatocellular iron and associated oxidative stress. Iron 170-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 2214784-6 1990 From the binding mode of ketoconazole to cytochrome P-450 and the present findings, it appears likely that the agent binds to a site which is different from that of steroids or pyridine nucleotides. Ketoconazole 25-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 41-57 2277761-7 1990 Cimetidine can bind to cytochrome P-450 covering the active haem group, the cytochrome proves to be of vital importance for hydroxylation reactions, involved in human steroidogenesis. Cimetidine 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 23-39 2223769-3 1990 Mutations to neutralize the basic charge at Arg 72 (R72Q) and to both neutralize and reverse the charge at Lys 344 (K344Q, K344E) resulted in alteration of NADH oxidation rates in the reconstituted physiological electron-transfer system, which is rate limited by putidaredoxin-cytochrome P-450cam electron transfer. Lysine 107-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 277-293 2277761-8 1990 Serum hydrocortisone concentrations will decrease when cytochrome P-450 becomes blocked. Hydrocortisone 6-20 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 2223769-3 1990 Mutations to neutralize the basic charge at Arg 72 (R72Q) and to both neutralize and reverse the charge at Lys 344 (K344Q, K344E) resulted in alteration of NADH oxidation rates in the reconstituted physiological electron-transfer system, which is rate limited by putidaredoxin-cytochrome P-450cam electron transfer. NAD 156-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 277-293 2223769-7 1990 Calculation of the cytochrome P-450cam electrostatic field revealed a patch of positive potential at the modeled cytochrome b5 interaction site lying directly above the nearest proximal approach to the buried heme prosthetic group. Heme 209-213 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 2233418-1 1990 The possibility that idiopathic Parkinson"s disease may be linked to a deficiency of an important detoxifying enzyme such as the cytochrome P450 enzyme bufuralol hydroxylase is examined. bufuralol 152-161 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 129-144 2200675-1 1990 Hepatocytes from adult and newborn humans were put into primary culture and exposed to phenobarbital, 3-methylcholanthrene, or rifampicin, three well-known inducers of cytochrome P-450 in animals. Rifampin 127-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 168-184 2133086-0 1990 Mechanism-based inactivation of human liver microsomal cytochrome P-450 IIIA4 by gestodene. Gestodene 81-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 55-71 2397214-7 1990 Potassium cation shows a bigger effect on the stability of cytochrome P-450 than spermine or cysteine, as revealed by a higher value of the pressure of half-inactivation, P1/2, and a bigger inactivation volume change. Potassium 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-75 2384159-4 1990 These results demonstrate the biosynthetic origin of the human kidney 11,12- and 14,15-epoxyeicosatrienoic acids and suggest a role for renal cytochrome P-450 in the bioactivation of endogenous pools of arachidonic acid. Arachidonic Acid 203-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-158 2397214-0 1990 The formation of cytochrome P-450 from cytochrome P-420 is promoted by spermine. Spermine 71-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 2397214-1 1990 This paper is concerned with camphor-bound bacterial cytochrome P-450 and processes that alter its spin-state equilibrium and influence its transition to the nonactive form, cytochrome P-420, as well as its renaturation to the native camphor-bound cytochrome P-450. Camphor 29-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 2397214-1 1990 This paper is concerned with camphor-bound bacterial cytochrome P-450 and processes that alter its spin-state equilibrium and influence its transition to the nonactive form, cytochrome P-420, as well as its renaturation to the native camphor-bound cytochrome P-450. Camphor 234-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 2375760-0 1990 Isolation of four forms of acetone-induced cytochrome P-450 in chicken liver by h.p.l.c. Acetone 27-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-59 2375760-2 1990 The purpose of this study was to purify and characterize the forms of cytochrome P-450 induced in chicken liver by acetone or ethanol. Acetone 115-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 2375760-2 1990 The purpose of this study was to purify and characterize the forms of cytochrome P-450 induced in chicken liver by acetone or ethanol. Ethanol 126-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-86 2375760-3 1990 Using high performance liquid ion-exchange chromatography, we were able to isolate at least four different forms of cytochrome P-450 which were induced by acetone in chicken liver. Acetone 155-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 116-132 2375760-8 1990 Another form of cytochrome P-450 induced by acetone is highly active in the hydroxylation of p-nitrophenol and in the conversion of acetaminophen to a reactive metabolite, similar to reactions catalysed by mammalian cytochrome P450IIE. Acetone 44-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 2375760-8 1990 Another form of cytochrome P-450 induced by acetone is highly active in the hydroxylation of p-nitrophenol and in the conversion of acetaminophen to a reactive metabolite, similar to reactions catalysed by mammalian cytochrome P450IIE. 4-nitrophenol 93-106 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 2375760-8 1990 Another form of cytochrome P-450 induced by acetone is highly active in the hydroxylation of p-nitrophenol and in the conversion of acetaminophen to a reactive metabolite, similar to reactions catalysed by mammalian cytochrome P450IIE. Acetaminophen 132-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-32 2375760-10 1990 A fourth form of cytochrome P-450 was identified whose N-terminal amino acid sequence and enzymic activities do not correspond to any mammalian cytochromes P-450 reported to be induced by acetone or ethanol. Acetone 188-195 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 2375760-10 1990 A fourth form of cytochrome P-450 was identified whose N-terminal amino acid sequence and enzymic activities do not correspond to any mammalian cytochromes P-450 reported to be induced by acetone or ethanol. Ethanol 199-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-33 2133086-1 1990 A series of 17 alpha-acetylenic steroids was examined with regard to ability to inactivate human liver microsomal cytochrome P-450 (P-450) IIA4, an enzyme involved in the oxidation of a number of drugs, carcinogens, and steroids, including estrogens and progestogens. alpha-acetylenic steroids 15-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 2133086-1 1990 A series of 17 alpha-acetylenic steroids was examined with regard to ability to inactivate human liver microsomal cytochrome P-450 (P-450) IIA4, an enzyme involved in the oxidation of a number of drugs, carcinogens, and steroids, including estrogens and progestogens. Steroids 32-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-130 2160283-0 1990 Cytochrome P-450 involvement in the NADPH-dependent lipid peroxidation in human placental mitochondria. NADP 36-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2168548-0 1990 Is the sigma opiate receptor a proadifen-sensitive subform of cytochrome P-450? Proadifen 31-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 62-78 2357863-1 1990 Because both quinidine and propranolol bind to the cytochrome P-450 responsible for the oxidation of debrisoquin, six healthy male subjects were studied to determine whether an interaction occurred between the two drugs and the pharmacodynamic consequences of that interaction. Quinidine 13-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 2357863-1 1990 Because both quinidine and propranolol bind to the cytochrome P-450 responsible for the oxidation of debrisoquin, six healthy male subjects were studied to determine whether an interaction occurred between the two drugs and the pharmacodynamic consequences of that interaction. Propranolol 27-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 2357863-1 1990 Because both quinidine and propranolol bind to the cytochrome P-450 responsible for the oxidation of debrisoquin, six healthy male subjects were studied to determine whether an interaction occurred between the two drugs and the pharmacodynamic consequences of that interaction. Debrisoquin 101-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 2161656-0 1990 Unusual hyperporphyrin spectrum by bis(glutathione dimethyl ester)-hemin complex, a model of cytochrome P-450-thiolate complexes. hyperporphyrin 8-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2161656-0 1990 Unusual hyperporphyrin spectrum by bis(glutathione dimethyl ester)-hemin complex, a model of cytochrome P-450-thiolate complexes. bis(glutathione dimethyl ester)-hemin 35-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2161656-0 1990 Unusual hyperporphyrin spectrum by bis(glutathione dimethyl ester)-hemin complex, a model of cytochrome P-450-thiolate complexes. thiolate 110-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2161656-2 1990 The structure of the complex was characterized by comparing their electronic absorption and electron spin resonance (ESR) spectra with those of thiolate adducts of cytochrome P-450 and their chemical model complexes. thiolate 144-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 164-180 2353556-1 1990 The inhibitory effects of pyridoglutethimide (3-ethyl-3-(4-pyridyl)piperidine-2,6-dione), an analogue of aminoglutethimide, on aromatase and other cytochrome P-450-dependent steroid-metabolizing enzymes were studied in vitro. rogletimide 26-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2353556-1 1990 The inhibitory effects of pyridoglutethimide (3-ethyl-3-(4-pyridyl)piperidine-2,6-dione), an analogue of aminoglutethimide, on aromatase and other cytochrome P-450-dependent steroid-metabolizing enzymes were studied in vitro. rogletimide 46-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2398265-5 1990 (a) Activities of the methyl cholanthrene-inducible forms of cytochrome P-450 were decreased compared to normal controls, whereas the activities of the phenobarbitone-inducible isozymes were relatively unaffected. Methylcholanthrene 22-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 61-77 2238705-8 1990 Type II binding is seen with inhibitors of mammalian cytochrome P-450 such as metyrapone, and with the imidazole antifungal agents such as clotrimazole. Metyrapone 78-88 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 2238705-8 1990 Type II binding is seen with inhibitors of mammalian cytochrome P-450 such as metyrapone, and with the imidazole antifungal agents such as clotrimazole. imidazole 103-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 2238705-12 1990 Avocado cytochrome P-450 catalysed the N-demethylation of N,N-dimethylaniline (17.1 nmol/min per nmol P-450) and p-chloro-N-methylaniline (13.1 nmol/min per nmol P-450), and the hydroxylation of lauric (dodecanoic) acid (1.1 nmol/min per nmol P-450). N,N-dimethylaniline 58-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2238705-12 1990 Avocado cytochrome P-450 catalysed the N-demethylation of N,N-dimethylaniline (17.1 nmol/min per nmol P-450) and p-chloro-N-methylaniline (13.1 nmol/min per nmol P-450), and the hydroxylation of lauric (dodecanoic) acid (1.1 nmol/min per nmol P-450). methylaniline 121-137 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2238705-12 1990 Avocado cytochrome P-450 catalysed the N-demethylation of N,N-dimethylaniline (17.1 nmol/min per nmol P-450) and p-chloro-N-methylaniline (13.1 nmol/min per nmol P-450), and the hydroxylation of lauric (dodecanoic) acid (1.1 nmol/min per nmol P-450). lauric (dodecanoic) acid 195-219 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 8-24 2353936-10 1990 These data suggest that mianserin is activated to a cytotoxic metabolite selectively by a constitutive form of human cytochrome P-450, whereas phenytoin, amitriptyline and imipramine are selectively activated by forms of mouse cytochrome P-450 which are induced by either phenobarbitone or beta-naphthoflavone. Mianserin 24-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-133 2353936-10 1990 These data suggest that mianserin is activated to a cytotoxic metabolite selectively by a constitutive form of human cytochrome P-450, whereas phenytoin, amitriptyline and imipramine are selectively activated by forms of mouse cytochrome P-450 which are induced by either phenobarbitone or beta-naphthoflavone. Mianserin 24-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 227-243 2372742-6 1990 The impairment on antipyrine disposition produced by piroxicam has been interpreted as a consequence of a reduction in the activity of hepatic microsomal drug-metabolizing enzymes, particularly the cytochrome P-450 system. Piroxicam 53-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 198-214 2231265-6 1990 Although the patterns of the inhibitory actions and the interaction of either imidazole drugs with cytochrome P-450 as 17 alpha-hydroxylase and C17,20-lyase were similar to those of KCZ, the inhibitory potencies and affinities for the cytochrome P-450 system decreased in the order of KCZ greater than MCZ greater than OZA greater than M-2 greater than M-1 greater than CIM. imidazole 78-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 2231265-8 1990 These results indicate that either imidazole drugs tested could inhibit a cytochrome P-450 enzyme C17,20-lyase mainly in testicular microsomes, and suggest that MCZ, a potent inhibitor subsequent to KCZ, induces a slight alteration in the testosterone biosynthesis in its clinical use. imidazole 35-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 2231265-8 1990 These results indicate that either imidazole drugs tested could inhibit a cytochrome P-450 enzyme C17,20-lyase mainly in testicular microsomes, and suggest that MCZ, a potent inhibitor subsequent to KCZ, induces a slight alteration in the testosterone biosynthesis in its clinical use. Miconazole 161-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 2231265-8 1990 These results indicate that either imidazole drugs tested could inhibit a cytochrome P-450 enzyme C17,20-lyase mainly in testicular microsomes, and suggest that MCZ, a potent inhibitor subsequent to KCZ, induces a slight alteration in the testosterone biosynthesis in its clinical use. Testosterone 239-251 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-90 2160283-1 1990 The NADPH-dependent lipid peroxidation in human placental mitochondria has been found to be inhibited strongly by amphenone B, aminoglutethimide and carbon monoxide, inhibitors of cytochrome P-450-mediated reactions, but was hardly affected by respiratory chain inhibitors. NADP 4-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 2160283-1 1990 The NADPH-dependent lipid peroxidation in human placental mitochondria has been found to be inhibited strongly by amphenone B, aminoglutethimide and carbon monoxide, inhibitors of cytochrome P-450-mediated reactions, but was hardly affected by respiratory chain inhibitors. amphenone B 114-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 2160283-1 1990 The NADPH-dependent lipid peroxidation in human placental mitochondria has been found to be inhibited strongly by amphenone B, aminoglutethimide and carbon monoxide, inhibitors of cytochrome P-450-mediated reactions, but was hardly affected by respiratory chain inhibitors. Aminoglutethimide 127-144 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 2160283-1 1990 The NADPH-dependent lipid peroxidation in human placental mitochondria has been found to be inhibited strongly by amphenone B, aminoglutethimide and carbon monoxide, inhibitors of cytochrome P-450-mediated reactions, but was hardly affected by respiratory chain inhibitors. Carbon Monoxide 149-164 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-196 2160283-2 1990 Cytochrome c, an exogenous electron acceptor which is known to compete with cytochrome P-450 for the reducing equivalents, showed an inhibitory effect on NADPH-dependent lipid peroxidation. NADP 154-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 76-92 2160283-7 1990 These data provide evidence that cytochrome P-450 is involved in NADPH-dependent mitochondrial lipid peroxidation. NADP 65-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 33-49 2160283-8 1990 It is suggested that superoxide liberated from cytochrome P-450, in combination with iron, may be responsible for initiation of NADPH-dependent lipid peroxidation in human placental mitochondria. Superoxides 21-31 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 2160283-8 1990 It is suggested that superoxide liberated from cytochrome P-450, in combination with iron, may be responsible for initiation of NADPH-dependent lipid peroxidation in human placental mitochondria. NADP 128-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 2376747-3 1990 Different function of the phenytoin-metabolizing cytochrome P450 subsystem was found in 6 patients, but in none of the controls. Phenytoin 26-35 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-64 1974199-5 1990 Only pretreatment of rabbits with rifampicin, which induces cytochrome P-450 form 3c (P-450IIIA6), significantly increased the microsomal hydroxylation of tolbutamide. Rifampin 34-44 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 1974199-5 1990 Only pretreatment of rabbits with rifampicin, which induces cytochrome P-450 form 3c (P-450IIIA6), significantly increased the microsomal hydroxylation of tolbutamide. Tolbutamide 155-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-76 1974199-11 1990 Hence, it appears that the animal model approach is not likely to be successful in the identification and characterization of the cytochrome P-450 form(s) metabolizing tolbutamide in humans. Tolbutamide 168-179 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 130-146 2160607-5 1990 (2) The catecholestrogen or diethylstilbestrol (DES) are oxidized to semiquinones and quinones by the peroxidatic activity of cytochrome P-450. Estrogens, Catechol 8-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-142 2160607-5 1990 (2) The catecholestrogen or diethylstilbestrol (DES) are oxidized to semiquinones and quinones by the peroxidatic activity of cytochrome P-450. Diethylstilbestrol 28-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-142 2160607-5 1990 (2) The catecholestrogen or diethylstilbestrol (DES) are oxidized to semiquinones and quinones by the peroxidatic activity of cytochrome P-450. Diethylstilbestrol 48-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-142 2160607-5 1990 (2) The catecholestrogen or diethylstilbestrol (DES) are oxidized to semiquinones and quinones by the peroxidatic activity of cytochrome P-450. semiquinones 69-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-142 2160607-5 1990 (2) The catecholestrogen or diethylstilbestrol (DES) are oxidized to semiquinones and quinones by the peroxidatic activity of cytochrome P-450. Quinones 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-142 2327982-0 1990 Differences in the cytochrome P-450 isoenzymes involved in the 2-hydroxylation of oestradiol and 17 alpha-ethinyloestradiol. Estradiol 82-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 2157443-0 1990 Inhibition of microsomal lipid peroxidation and cytochrome P-450-catalyzed reactions by beta-lapachone and related naphthoquinones. beta-lapachone 88-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 2344166-10 1990 Nonspecific inhibition of cytochrome P-450 was ruled out since erythromycin N demethylation (cytochrome P-450 mediated) was unaffected in the presence of enoxacin. Erythromycin 63-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 93-109 2344166-12 1990 We conclude that some quinolones are potent and selective inhibitors of specific isozymes of human cytochrome P-450 that are responsible for theophylline metabolism. Quinolones 22-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 2344166-12 1990 We conclude that some quinolones are potent and selective inhibitors of specific isozymes of human cytochrome P-450 that are responsible for theophylline metabolism. Theophylline 141-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 99-115 2157443-0 1990 Inhibition of microsomal lipid peroxidation and cytochrome P-450-catalyzed reactions by beta-lapachone and related naphthoquinones. Naphthoquinones 115-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-64 2327982-0 1990 Differences in the cytochrome P-450 isoenzymes involved in the 2-hydroxylation of oestradiol and 17 alpha-ethinyloestradiol. 17 alpha-ethinyloestradiol 97-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-35 2157443-4 1990 These observations support the hypothesis that, in the micromolar concentration range, o-naphthoquinones inhibit microsomal lipid peroxidation and cytochrome P-450-catalyzed reactions, by diverting reducing equivalents from NADPH to dioxygen. 1,2-naphthoquinone 87-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2378912-0 1990 [Chemical modification of cholesterol-hydroxylating cytochrome P-450 from adrenal cortex mitochondria with diethylpyrocarbonate]. Cholesterol 26-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 2157443-4 1990 These observations support the hypothesis that, in the micromolar concentration range, o-naphthoquinones inhibit microsomal lipid peroxidation and cytochrome P-450-catalyzed reactions, by diverting reducing equivalents from NADPH to dioxygen. NADP 224-229 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2157443-4 1990 These observations support the hypothesis that, in the micromolar concentration range, o-naphthoquinones inhibit microsomal lipid peroxidation and cytochrome P-450-catalyzed reactions, by diverting reducing equivalents from NADPH to dioxygen. Oxygen 233-241 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-163 2340358-2 1990 The samples analyzed consisted of the chlorophenols resulting from the cytochrome P-450 mediated oxidation of isotopic mixtures of chlorobenzenes, and were analyzed on a VG-7070 double-focusing mass spectrometer. Chlorophenols 38-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 2340358-2 1990 The samples analyzed consisted of the chlorophenols resulting from the cytochrome P-450 mediated oxidation of isotopic mixtures of chlorobenzenes, and were analyzed on a VG-7070 double-focusing mass spectrometer. Chlorobenzenes 131-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-87 2378912-0 1990 [Chemical modification of cholesterol-hydroxylating cytochrome P-450 from adrenal cortex mitochondria with diethylpyrocarbonate]. Diethyl Pyrocarbonate 107-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 2180560-5 1990 Both rat cytochrome P-450 beta NF-B (P-450 IA1) and P-450ISF-G (P-450 IA2) inactivated 1,3-dinitropyrene, with the former being more effective. 1,3-dinitropyrene 87-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 9-25 2180560-0 1990 Inactivation of 1,3-, 1,6-, and 1,8-dinitropyrene by cytochrome P-450 enzymes in human and rat liver microsomes. 1,3-, 1,6-, and 1,8-dinitropyrene 16-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 53-69 2182022-6 1990 It is likely that the generation of the radical proceeds via a ferryl intermediate, as in the proposed mechanisms for cytochrome P-450 and the peroxidases. radical 40-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-134 2180560-6 1990 Correlation studies done with liver microsomes prepared from variously treated rats and immunoinhibition studies suggest that cytochrome P-450 beta NF-B and P-450ISF-G are both involved in the detoxication of all three of the dinitropyrenes in rat liver microsomes. dinitropyrenes 226-240 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-142 2180560-8 1990 Correlation analysis and inhibition studies with 7,8-benzoflavone and antibodies indicate that human cytochrome P-450 enzymes in the IA family are most effective in detoxicating this compound; the contribution of cytochrome P-450PA (P-450 IA2, the phenacetin O-deethylase) is deemed more important, but a role for the small amount of cytochrome P1-450 (P-450 IA1) in the liver cannot be ruled out. alpha-naphthoflavone 49-65 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 101-117 2180560-11 1990 Thus, distinct human cytochrome P-450 enzymes are involved in the detoxication of different dinitropyrene congeners, and the situation appears to contrast with that in rat liver. DINITROPYRENE 92-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-37 2155225-1 1990 Role of aldehyde dehydrogenase and leukotriene B4 omega-hydroxylase (cytochrome P-450LTB omega) in leukotriene B4 omega-oxidation. Leukotrienes 35-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-94 2155225-4 1990 This activity has been ascribed to LTB4 omega-hydroxylase (cytochrome P-450LTB omega), a conclusion supported by our finding of the reversal of carbon monoxide inhibition by 450 nm light and by competitive inhibition studies. Carbon Monoxide 144-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-84 2155225-5 1990 The oxidation of 20-oxo-LTB4 to 20-carboxy-LTB4 is also catalyzed by microsomes fortified with 1 mM NAD+, and this activity is not affected by cytochrome P-450LTB omega inhibitors. 20-aldehyde leukotriene B4 17-28 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-168 2155225-5 1990 The oxidation of 20-oxo-LTB4 to 20-carboxy-LTB4 is also catalyzed by microsomes fortified with 1 mM NAD+, and this activity is not affected by cytochrome P-450LTB omega inhibitors. 20-carboxyleukotriene B4 32-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-168 2310416-6 1990 Covalent binding of TAI, as that of TA, depends on cytochrome P-450-dependent monooxygenases and is almost completely inhibited in the presence of sulfur containing nucleophiles such as glutathione, cysteine or cyteamine. Ticrynafen 20-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 2328827-1 1990 The cytochrome P-450-dependent aromatase pathway utilizes the androgens testosterone (T) and androstenedione, as substrates for estrogen formation. Testosterone 72-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2328827-1 1990 The cytochrome P-450-dependent aromatase pathway utilizes the androgens testosterone (T) and androstenedione, as substrates for estrogen formation. Androstenedione 93-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2310416-6 1990 Covalent binding of TAI, as that of TA, depends on cytochrome P-450-dependent monooxygenases and is almost completely inhibited in the presence of sulfur containing nucleophiles such as glutathione, cysteine or cyteamine. Sulfur 147-153 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 2310416-6 1990 Covalent binding of TAI, as that of TA, depends on cytochrome P-450-dependent monooxygenases and is almost completely inhibited in the presence of sulfur containing nucleophiles such as glutathione, cysteine or cyteamine. Glutathione 186-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 2310416-6 1990 Covalent binding of TAI, as that of TA, depends on cytochrome P-450-dependent monooxygenases and is almost completely inhibited in the presence of sulfur containing nucleophiles such as glutathione, cysteine or cyteamine. cyteamine 211-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 51-67 2310416-7 1990 These results show that 5-OHTA, which has been reported as the major metabolite of TA in vivo in humans, is formed by liver microsomes by a cytochrome P-450-dependent reaction. 5-ohta 24-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 2310416-7 1990 These results show that 5-OHTA, which has been reported as the major metabolite of TA in vivo in humans, is formed by liver microsomes by a cytochrome P-450-dependent reaction. Ticrynafen 28-30 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 2314392-0 1990 Mechanisms of activation of phenacetin to reactive metabolites by cytochrome P-450: a theoretical study involving radical intermediates. Phenacetin 28-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 2369956-0 1990 [Immunomodulators with an 8-azasteroid structure as inducers of liver cytochrome P-450]. 8-azasteroid 26-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-87 2369956-1 1990 Two structural analogues of D-homo-8-azasteroids, both an immunostimulant and an immunodepressant, are inductors of the liver cytochrome P-450 in animals. d-homo-8-azasteroids 28-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 126-142 2312782-1 1990 Quinidine has been reported to be a potent inhibitor of a specific isozyme of cytochrome P-450 (P-450db 1) that is responsible for the metabolism of a select group of drugs. Quinidine 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-94 2312782-2 1990 In order to investigate the potential for quinidine to inhibit other isozymes of cytochrome P-450 and to assess whether or not P-450db 1 plays any role in antipyrine metabolism, we studied the effects of quinidine pretreatment on the pharmacokinetics and metabolism of antipyrine in six healthy, male volunteers. Quinidine 42-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 2314392-1 1990 The cytochrome P-450-mediated activation of phenacetin (PHEN) to reactive intermediates by two hypothetical mechanisms has been studied by use of SV 6-31G ab initio energy and spin distribution calculations. Phenacetin 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2314392-1 1990 The cytochrome P-450-mediated activation of phenacetin (PHEN) to reactive intermediates by two hypothetical mechanisms has been studied by use of SV 6-31G ab initio energy and spin distribution calculations. Phenacetin 56-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2314392-2 1990 In our calculations, the cytochrome P-450 enzyme system has been substituted by a singlet oxygen atom in order to reduce the computational efforts and to fulfill the requirements as to spin conservation. Singlet Oxygen 82-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-41 2310385-1 1990 Arachidonic acid (AA) can be metabolized in endothelial cells (EC) to a series of epoxides via cytochrome P-450 epoxygenase with 14,15 epoxyeicosatrienoic acid (14,15-EET) as the major product. Arachidonic Acid 0-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 2310385-1 1990 Arachidonic acid (AA) can be metabolized in endothelial cells (EC) to a series of epoxides via cytochrome P-450 epoxygenase with 14,15 epoxyeicosatrienoic acid (14,15-EET) as the major product. Epoxy Compounds 82-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 2310385-1 1990 Arachidonic acid (AA) can be metabolized in endothelial cells (EC) to a series of epoxides via cytochrome P-450 epoxygenase with 14,15 epoxyeicosatrienoic acid (14,15-EET) as the major product. 14,15-epoxy-5,8,11-eicosatrienoic acid 129-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 2310385-1 1990 Arachidonic acid (AA) can be metabolized in endothelial cells (EC) to a series of epoxides via cytochrome P-450 epoxygenase with 14,15 epoxyeicosatrienoic acid (14,15-EET) as the major product. 14,15-epoxy-5,8,11-eicosatrienoic acid 161-170 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-111 2328938-2 1990 The CYP21B gene encodes an adrenal microsomal cytochrome P-450, which is specific for steroid 21-hydroxylation (P450c21). Steroids 86-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-62 2182119-0 1990 Tyrosine-96 as a natural spectroscopic probe of the cytochrome P-450cam active site. Tyrosine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-68 2155989-4 1990 These results suggest that the dose-dependent dual effect of silibinin on corticosteroid secretion may be attributed to corresponding changes in the activities of cytochrome P-450 enzymes, and that stimulation of ACTH-induced corticosteroidogenesis by silibinin is presumably due to the antioxidant property of the drug. Silybin 61-70 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 163-179 2298305-0 1990 Heme maintains catalytically active structure of cytochrome P-450. Heme 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-65 1967940-1 1990 Metyrapone, an inhibitor of cytochrome P-450-dependent monooxygenases, enhanced the induction of tyrosine aminotransferase by dexamethasone in primary cultures of hepatocytes, while it had no effect on the basal level of the enzyme activity in the absence of the hormone. Metyrapone 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 1967940-1 1990 Metyrapone, an inhibitor of cytochrome P-450-dependent monooxygenases, enhanced the induction of tyrosine aminotransferase by dexamethasone in primary cultures of hepatocytes, while it had no effect on the basal level of the enzyme activity in the absence of the hormone. Dexamethasone 126-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-44 2154220-0 1990 A possible role of cAMP dependent phosphorylation of hepatic microsomal cytochrome P450: a mechanism to increase lipid peroxidation in response to hormone. Cyclic AMP 19-23 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-87 2154220-2 1990 cAMP dependent phosphorylation of cytochrome P450 was found to increase the NADPH dependent production of malondialdehyde (lipid peroxidation) by about 30%. Cyclic AMP 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 2154220-2 1990 cAMP dependent phosphorylation of cytochrome P450 was found to increase the NADPH dependent production of malondialdehyde (lipid peroxidation) by about 30%. NADP 76-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 1967940-4 1990 The dexamethasone activity amplification by metyrapone could be the consequence of a modulation of the glucocorticoid biotransformations due to the cytochrome P-450 inhibitor. Dexamethasone 4-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 2298305-1 1990 Treatment of purified cytochrome P-450 LM2 and its liposome-bound form with hydrogen peroxide led to complete destruction of the P-450 heme. Hydrogen Peroxide 76-93 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 1967940-4 1990 The dexamethasone activity amplification by metyrapone could be the consequence of a modulation of the glucocorticoid biotransformations due to the cytochrome P-450 inhibitor. Metyrapone 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-164 2154220-2 1990 cAMP dependent phosphorylation of cytochrome P450 was found to increase the NADPH dependent production of malondialdehyde (lipid peroxidation) by about 30%. Malondialdehyde 106-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 34-49 2154220-3 1990 The cytochrome P450 inhibitor cyanide abolished this activity. Cyanides 30-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 2298305-1 1990 Treatment of purified cytochrome P-450 LM2 and its liposome-bound form with hydrogen peroxide led to complete destruction of the P-450 heme. Heme 135-139 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 22-38 2154220-4 1990 The presence of spermine decreased the cytochrome P450 dependent lipid peroxidation in non-phosphorylated microsomes, phosphorylation partially reversed this effect. Spermine 16-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 39-54 2298305-2 1990 The apoenzyme thus produced could be reconstituted to catalytically active cytochrome P-450 by incubation with hemin, the reconstitution efficiency being 50% for the soluble enzyme and 80% for the liposome-bound enzyme. Hemin 111-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 2154220-7 1990 It is hypothesized that phosphorylation of cytochrome P450 alters the interaction between the components of the cytochrome P450 system, which may enhance production of free radical species, initiating lipid peroxidation. Free Radicals 168-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 2154220-7 1990 It is hypothesized that phosphorylation of cytochrome P450 alters the interaction between the components of the cytochrome P450 system, which may enhance production of free radical species, initiating lipid peroxidation. Free Radicals 168-180 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-127 2371836-0 1990 "In vitro" cyclosporin and methylprednisolone interaction with cytochrome P-450. Cyclosporine 11-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 2371836-0 1990 "In vitro" cyclosporin and methylprednisolone interaction with cytochrome P-450. Methylprednisolone 27-45 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 2344458-1 1990 An electrochemical system of cytochrome P-450 reduction in the presence of the water-soluble redox carrier methylviologen has been developed. Water 79-84 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-45 2156510-0 1990 Antibodies against rabbit omega-hydroxylases of prostaglandins and fatty acids cross-react with leukotriene B4 omega-hydroxylase in human neutrophils (cytochrome P-450LTB omega). Prostaglandins 48-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-176 2156510-0 1990 Antibodies against rabbit omega-hydroxylases of prostaglandins and fatty acids cross-react with leukotriene B4 omega-hydroxylase in human neutrophils (cytochrome P-450LTB omega). Fatty Acids 67-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 151-176 2344452-0 1990 [The role of heme in formation of the native structure of cytochrome P-450 LM2]. Heme 13-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 58-74 24487082-4 1990 Other drugs, such as chloramphenicol, propoxyphene, verapamil, and viloxazine, inhibit cytochrome P-450. Chloramphenicol 21-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-103 24487082-4 1990 Other drugs, such as chloramphenicol, propoxyphene, verapamil, and viloxazine, inhibit cytochrome P-450. Dextropropoxyphene 38-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-103 24487082-4 1990 Other drugs, such as chloramphenicol, propoxyphene, verapamil, and viloxazine, inhibit cytochrome P-450. Verapamil 52-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-103 24487082-4 1990 Other drugs, such as chloramphenicol, propoxyphene, verapamil, and viloxazine, inhibit cytochrome P-450. Viloxazine 67-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-103 2344458-2 1990 In this system cytochrome P-450 effectuates a steady-state demethylation of dimethylaniline and hydroxylation of aniline. N,N-dimethylaniline 76-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 2344452-1 1990 The role of heme in the formation of cytochrome P-450 native structure was investigated. Heme 12-16 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-53 2344454-6 1990 Incubation of the immobilized cytochrome P-450 LM 2 with sonicated liposomes composed of various phospholipids did not result in oligomer dissociation and protein translocation from the matrix to the lipid phase, although the catalytic activity of the immobilized cytochrome significantly increased in the presence of liposomes. Phospholipids 97-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-46 2344458-2 1990 In this system cytochrome P-450 effectuates a steady-state demethylation of dimethylaniline and hydroxylation of aniline. aniline 84-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-31 2322954-0 1990 The role of hydrophobicity and electronic factors in regulating alcohol inhibition of cytochrome P-450-mediated aniline hydroxylation. Alcohols 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 2131820-7 1990 These results are interpreted in terms of an electron/proton/electron transfer mechanism previously postulated for the oxidation of other dihydropyridines by cytochrome P-450 enzymes and model one-electron acceptors and argue against a mechanism involving hydrogen atom abstraction from nifedipine. Dihydropyridines 138-154 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-174 2131820-7 1990 These results are interpreted in terms of an electron/proton/electron transfer mechanism previously postulated for the oxidation of other dihydropyridines by cytochrome P-450 enzymes and model one-electron acceptors and argue against a mechanism involving hydrogen atom abstraction from nifedipine. Hydrogen 256-264 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-174 2131820-7 1990 These results are interpreted in terms of an electron/proton/electron transfer mechanism previously postulated for the oxidation of other dihydropyridines by cytochrome P-450 enzymes and model one-electron acceptors and argue against a mechanism involving hydrogen atom abstraction from nifedipine. Nifedipine 287-297 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 158-174 2403855-0 1990 Metabolic deactivation of furylfuramide by cytochrome P450 in human and rat liver microsomes. Furylfuramide 26-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 2322954-0 1990 The role of hydrophobicity and electronic factors in regulating alcohol inhibition of cytochrome P-450-mediated aniline hydroxylation. aniline 112-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 2322954-1 1990 The role of hydrophobicity and electronic factors in regulating alcohol inhibition of cytochrome P-450-mediated aniline p-hydroxylation has been investigated by the formulation of quantitative structure-activity relationships. Alcohols 64-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 2322954-1 1990 The role of hydrophobicity and electronic factors in regulating alcohol inhibition of cytochrome P-450-mediated aniline p-hydroxylation has been investigated by the formulation of quantitative structure-activity relationships. aniline 112-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 86-102 2322954-6 1990 Similarities are found between alcohol inhibition and the binding of alkyl amines to cytochrome P-450, suggesting that alcohols may bind to the amine binding site. Alcohols 31-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 2322954-6 1990 Similarities are found between alcohol inhibition and the binding of alkyl amines to cytochrome P-450, suggesting that alcohols may bind to the amine binding site. alkyl amines 69-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 2322954-6 1990 Similarities are found between alcohol inhibition and the binding of alkyl amines to cytochrome P-450, suggesting that alcohols may bind to the amine binding site. Alcohols 119-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 2322954-6 1990 Similarities are found between alcohol inhibition and the binding of alkyl amines to cytochrome P-450, suggesting that alcohols may bind to the amine binding site. Amines 75-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-101 2265588-0 1990 [The role of cytochrome P-450 in detoxifying the thioethynyl esters of monothiophosphoric acids]. thioethynyl esters 49-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 2264925-8 1990 In the kidney, cis-platinum appears to be the most effective inducer of cytochrome P-450, whereby the biotransformation of the prostaglandins and fatty acids could be altered. Cisplatin 15-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 2264925-8 1990 In the kidney, cis-platinum appears to be the most effective inducer of cytochrome P-450, whereby the biotransformation of the prostaglandins and fatty acids could be altered. Prostaglandins 127-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 2264925-8 1990 In the kidney, cis-platinum appears to be the most effective inducer of cytochrome P-450, whereby the biotransformation of the prostaglandins and fatty acids could be altered. Fatty Acids 146-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 72-88 2264925-9 1990 The spectrum of the effects of cis-platinum on cytochrome P-450-dependent drug metabolism and steroid hydroxylation activity mimic those produced by hypophysectomy and are for the most part reversed by the anterior pituitary hormones. Cisplatin 31-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 47-63 2265588-0 1990 [The role of cytochrome P-450 in detoxifying the thioethynyl esters of monothiophosphoric acids]. thiophosphoric acid 71-95 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-29 2250572-1 1990 The administration of doxorubicin, an anti-tumor antibiotic, to rodents resulted in an increase in heme oxygenase activity and a decrease in delta-aminolevulinate (ALA) synthase activity and in cellular heme and cytochrome P450 content in liver. Doxorubicin 22-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 212-227 1699849-3 1990 In addition, the interaction of lindane with the liver tissue results in the induction of the microsomal cytochrome P-450 system, together with enhanced rates of superoxide radical generation and a significant increase in indicators of lipid peroxidation. Hexachlorocyclohexane 32-39 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 1693736-8 1990 The only significant drug interactions with felodipine occur with inducers and inhibitors of the cytochrome P-450 system, which is responsible for the metabolism of felodipine. Felodipine 44-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 1693736-8 1990 The only significant drug interactions with felodipine occur with inducers and inhibitors of the cytochrome P-450 system, which is responsible for the metabolism of felodipine. Felodipine 165-175 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-113 2250572-2 1990 Sn-protoporphyrin, a potent inhibitor of heme degradation both in vitro and in vivo, when administered to rodents prior to doxorubicin, mitigates the drug-induced toxic actions which are reflected by the drug-induced decreases of both cellular heme and cytochrome P450 content. tin protoporphyrin IX 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 253-268 2273938-7 1990 Levels of cytochrome P-450 IIIA4 vary widely among individuals and can explain the variation in rates of 17 alpha-ethynylestradiol 2-hydroxylation. Ethinyl Estradiol 105-130 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-26 2250572-3 1990 Sn-protoporphyrin thus provides a pharmacological means of protecting against the toxic effects of doxorubicin and other drugs which enhance heme oxygenase activity and thus decrease cellular heme and cytochrome P450 content in vivo. tin protoporphyrin IX 0-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-216 2250572-3 1990 Sn-protoporphyrin thus provides a pharmacological means of protecting against the toxic effects of doxorubicin and other drugs which enhance heme oxygenase activity and thus decrease cellular heme and cytochrome P450 content in vivo. Doxorubicin 99-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 201-216 2273938-9 1990 Mechanism-based inactivation of cytochrome P-450 IIIA4 can be seen with 17 alpha-ethynylestradiol and other 17 alpha-acetylenic steroids, and the progestogen gestodene appears to be unusually active in this regard. Ethinyl Estradiol 72-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 2151997-0 1990 Cytochrome P-450s as toxicogenic catalysts: the influence of dehydroepiandrosterone. Dehydroepiandrosterone 61-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-16 2273938-9 1990 Mechanism-based inactivation of cytochrome P-450 IIIA4 can be seen with 17 alpha-ethynylestradiol and other 17 alpha-acetylenic steroids, and the progestogen gestodene appears to be unusually active in this regard. alpha-acetylenic steroids 111-136 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-48 2273938-10 1990 Other unknown factors may also modulate levels of cytochrome P-450 IIIA4 and its ability to catalyze 17 alpha-ethynylestradiol 2-hydroxylation. Ethinyl Estradiol 101-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-66 2134673-4 1990 Interestingly, the clofibrate receptor, believed to be involved in clofibrate induction of certain isozymes of cytochrome P-450, has recently been shown to be a member of the steroid receptor supergene family and it is conceivable that other P-450 inducers might act via as yet uncharacterized receptors belonging to the same gene family. Clofibrate 19-29 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 111-127 2134683-3 1990 The cytochrome P-450 enzymes are also responsible for the metabolism of endogenous substrates including fatty acids, prostaglandins and all classes of steroids. Fatty Acids 104-115 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2134683-3 1990 The cytochrome P-450 enzymes are also responsible for the metabolism of endogenous substrates including fatty acids, prostaglandins and all classes of steroids. Prostaglandins 117-131 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 2122185-0 1990 Ocular cytochrome P-450 metabolism of arachidonate: synthesis and bioassay. Arachidonic Acid 38-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-23 2181486-2 1990 Ethanol is also oxidized in liver microsomes by an ethanol-inducible cytochrome P-450 (P-450IIE1) which contributes to ethanol metabolism and tolerance, and activates xenobiotics to toxic radicals thereby explaining increased vulnerability of the heavy drinker to industrial solvents, anesthetic agents, commonly prescribed drugs, over-the-counter analgesics, chemical carcinogens and even nutritional factors such as vitamin A. Ethanol 0-7 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 2181486-2 1990 Ethanol is also oxidized in liver microsomes by an ethanol-inducible cytochrome P-450 (P-450IIE1) which contributes to ethanol metabolism and tolerance, and activates xenobiotics to toxic radicals thereby explaining increased vulnerability of the heavy drinker to industrial solvents, anesthetic agents, commonly prescribed drugs, over-the-counter analgesics, chemical carcinogens and even nutritional factors such as vitamin A. Ethanol 51-58 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 2181486-2 1990 Ethanol is also oxidized in liver microsomes by an ethanol-inducible cytochrome P-450 (P-450IIE1) which contributes to ethanol metabolism and tolerance, and activates xenobiotics to toxic radicals thereby explaining increased vulnerability of the heavy drinker to industrial solvents, anesthetic agents, commonly prescribed drugs, over-the-counter analgesics, chemical carcinogens and even nutritional factors such as vitamin A. Ethanol 119-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 2181486-2 1990 Ethanol is also oxidized in liver microsomes by an ethanol-inducible cytochrome P-450 (P-450IIE1) which contributes to ethanol metabolism and tolerance, and activates xenobiotics to toxic radicals thereby explaining increased vulnerability of the heavy drinker to industrial solvents, anesthetic agents, commonly prescribed drugs, over-the-counter analgesics, chemical carcinogens and even nutritional factors such as vitamin A. Vitamin A 418-427 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 69-85 2181493-4 1990 Currently available in vivo and in vitro data suggest that nitrosamines may be metabolized by cytochrome P-450, prostaglandin endoperoxide synthetase, or monoamine oxidases. Nitrosamines 59-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 94-110 2134683-3 1990 The cytochrome P-450 enzymes are also responsible for the metabolism of endogenous substrates including fatty acids, prostaglandins and all classes of steroids. Steroids 151-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-20 20837414-10 1990 These results indicate that isomers of Cl-DBFs induce effectively 3-methylcholanthrene-inducible type of cytochrome P-450. cl-dbfs 39-46 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 2162582-2 1990 To exemplify the first-group reactions, cytochrome P-450 was studied, which is capable of generating different active forms of oxygen during the catalytic cycle. Oxygen 127-133 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 40-56 2162582-4 1990 In the course of the reaction, cytochrome P-450 also became inactivated under the effect of active oxygen. Oxygen 99-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-47 20837414-10 1990 These results indicate that isomers of Cl-DBFs induce effectively 3-methylcholanthrene-inducible type of cytochrome P-450. Methylcholanthrene 66-86 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 105-121 33809117-3 2021 A clearer understanding of CYP expression is important in the pathogenesis of breast cancer as several isoforms play critical roles in metabolising steroid hormones and xenobiotics that contribute to the genesis of breast cancer. Steroids 148-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 2220168-1 1990 Caffeine is mainly metabolized by 3-methyl-cholanthrene-inducible cytochrome P-450, whereas metamizol (Analgin) is probably mainly metabolized by phenobarbital inducible cytochromes P-450. Caffeine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 2220168-1 1990 Caffeine is mainly metabolized by 3-methyl-cholanthrene-inducible cytochrome P-450, whereas metamizol (Analgin) is probably mainly metabolized by phenobarbital inducible cytochromes P-450. Methylcholanthrene 34-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-82 2220168-2 1990 Therefore the elimination of caffeine from serum and the amount of the main metabolites of metamizol excreted into urine reflect the activity of these two cytochrome P-450 families. Caffeine 29-37 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 2220168-2 1990 Therefore the elimination of caffeine from serum and the amount of the main metabolites of metamizol excreted into urine reflect the activity of these two cytochrome P-450 families. Dipyrone 91-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-171 33973477-5 2021 The most studied drug categories were anticonvulsants and selective serotonin reuptake inhibitors associated with human leukocyte antigen and cytochrome P450 genes (HLA-A, HLA-B, CYP2C9, CYP2D6, CYP2C19), classified as critically low quality/low quality. Serotonin 68-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 142-157 34843882-7 2022 Further study showed inhibitory effect of HGK on CYP enzymes was weaker than that of diosmetin, which may be related to the substitution of hydroxyl and methoxy in the A and B rings of the flavone skeleton. flavone 189-196 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 49-52 23859801-4 2013 Lacosamide has a well-characterized and favorable pharmacokinetic profile, including a fast absorption rate, minimal or no interaction with cytochrome P-450 izoenzymes, and a low potential for drug-drug interactions. Lacosamide 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 140-156 34847475-0 2022 The impact of individual human cytochrome P450 enzymes on oxidative metabolism of anticancer drug lenvatinib. lenvatinib 98-108 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 34410044-0 2022 Translational aspects of cytochrome P450-mediated drug-drug interactions: a case study with clopidogrel. Clopidogrel 92-103 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 25-40 34410044-10 2022 This MiniReview provides an overview of translational approaches to study CYP-mediated DDIs, going beyond regulatory DDI guidelines, and an illustrative case study of how the DDI potential of clopidogrel was unveiled by combining these different methods. Clopidogrel 192-203 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-77 34861484-2 2022 However, cancer drug resistance greatly mitigates the cure rates of tumors, and cytochrome P450 (CYP450) plays an important role in the development of cisplatin resistance. Cisplatin 151-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 34780725-0 2022 Dabrafenib inhibits ABCG2 and cytochrome P450 isoenzymes; potential implications for combination anticancer therapy. dabrafenib 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 30-45 34352543-0 2022 Computational insight into biotransformation of halophenols by cytochrome P450: Mechanism and reactivity for epoxidation. halophenols 48-59 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 34352543-2 2022 Previous experimental studies have shown that XPs can be catalytically transformed into epoxides and haloquinones by cytochrome P450 enzymes (CYPs). Epoxy Compounds 88-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-132 34352543-2 2022 Previous experimental studies have shown that XPs can be catalytically transformed into epoxides and haloquinones by cytochrome P450 enzymes (CYPs). haloquinone 101-113 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-132 34426286-1 2022 OBJECTIVE: Bisphenol A (BPA), a common endocrine disrupter, can be activated by cytochrome P450 (CYP) metabolizing enzymes and might influence the development of breast cancer (BC). bisphenol A 11-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 34426286-1 2022 OBJECTIVE: Bisphenol A (BPA), a common endocrine disrupter, can be activated by cytochrome P450 (CYP) metabolizing enzymes and might influence the development of breast cancer (BC). bisphenol A 11-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 34426286-1 2022 OBJECTIVE: Bisphenol A (BPA), a common endocrine disrupter, can be activated by cytochrome P450 (CYP) metabolizing enzymes and might influence the development of breast cancer (BC). bisphenol A 24-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 34426286-1 2022 OBJECTIVE: Bisphenol A (BPA), a common endocrine disrupter, can be activated by cytochrome P450 (CYP) metabolizing enzymes and might influence the development of breast cancer (BC). bisphenol A 24-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 97-100 34426286-2 2022 We hypothesized that BPA could interact with CYP genes, synergistically contributing to the BC risk. bisphenol A 21-24 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 45-48 34780725-8 2022 Overall, our data confirm dabrafenib as a drug frequently and potently interacting with ABC transporters and CYP isoenzymes. dabrafenib 26-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 109-112 34493602-8 2021 These data suggest that cannabinoids and major THC metabolites are able to inhibit the activities of multiple CYP enzymes, and basic static modelling of these data suggest the possibility of pharmacokinetic interactions between these cannabinoids and xenobiotics extensively metabolized by CYP2B6, CYP2C9 and CYP2D6. Cannabinoids 24-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 34806536-0 2022 Salinomycin induces cell cycle arrest and apoptosis and modulates hepatic cytochrome P450 mRNA expression in HepG2/C3a cells. salinomycin 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 74-89 34806536-8 2022 This study also demonstrated the ability of SAL in modulating hepatic cytochrome P450 (CYP) mRNA expression, such that SAL caused the upregulation of CYP1A members and CYP3A5; and downregulation of CYP3A4. salinomycin 44-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 34806536-8 2022 This study also demonstrated the ability of SAL in modulating hepatic cytochrome P450 (CYP) mRNA expression, such that SAL caused the upregulation of CYP1A members and CYP3A5; and downregulation of CYP3A4. salinomycin 44-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 34806536-8 2022 This study also demonstrated the ability of SAL in modulating hepatic cytochrome P450 (CYP) mRNA expression, such that SAL caused the upregulation of CYP1A members and CYP3A5; and downregulation of CYP3A4. salinomycin 119-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 70-85 34806536-8 2022 This study also demonstrated the ability of SAL in modulating hepatic cytochrome P450 (CYP) mRNA expression, such that SAL caused the upregulation of CYP1A members and CYP3A5; and downregulation of CYP3A4. salinomycin 119-122 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 87-90 34877801-1 2022 This study was designed to evaluate the effects of cenobamate, an antiseizure medication for focal seizures, on the pharmacokinetics (PK) of cytochrome P450 probes (bupropion, CYP2B6; midazolam, CYP3A4/5; warfarin, CYP2C9; omeprazole, CYP2C19) in healthy subjects. Bupropion 165-174 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-156 34877801-1 2022 This study was designed to evaluate the effects of cenobamate, an antiseizure medication for focal seizures, on the pharmacokinetics (PK) of cytochrome P450 probes (bupropion, CYP2B6; midazolam, CYP3A4/5; warfarin, CYP2C9; omeprazole, CYP2C19) in healthy subjects. Midazolam 184-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 141-156 34363188-1 2021 BACKGROUND: Hepatic impairment can impact apixaban pharmacokinetics and pharmacodynamics by decreasing cytochrome P450-mediated metabolism and factor X production. apixaban 42-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 103-118 34493602-8 2021 These data suggest that cannabinoids and major THC metabolites are able to inhibit the activities of multiple CYP enzymes, and basic static modelling of these data suggest the possibility of pharmacokinetic interactions between these cannabinoids and xenobiotics extensively metabolized by CYP2B6, CYP2C9 and CYP2D6. Dronabinol 47-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 34493602-8 2021 These data suggest that cannabinoids and major THC metabolites are able to inhibit the activities of multiple CYP enzymes, and basic static modelling of these data suggest the possibility of pharmacokinetic interactions between these cannabinoids and xenobiotics extensively metabolized by CYP2B6, CYP2C9 and CYP2D6. Cannabinoids 234-246 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 34493602-9 2021 Significance Statement Major cannabinoids and their metabolites found in the plasma of cannabis users inhibit several CYP enzymes, including CYP2B6, CYP2C9, and CYP2D6. Cannabinoids 29-41 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 118-121 34648192-9 2022 CONCLUSION: CYP1A2 is the major CYP for the conversion of 4-MAA to 4-AA and 4-FAA. 4-maa 58-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 34848204-0 2022 Cytochrome P450-derived fatty acid epoxides and diols in angiogenesis and stem cell biology. Fatty Acids 24-34 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34848204-0 2022 Cytochrome P450-derived fatty acid epoxides and diols in angiogenesis and stem cell biology. Epoxy Compounds 35-43 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34848204-1 2022 Cytochrome P450 (CYP) enzymes are frequently referred to as the third pathway for the metabolism of arachidonic acid. Arachidonic Acid 100-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34848204-1 2022 Cytochrome P450 (CYP) enzymes are frequently referred to as the third pathway for the metabolism of arachidonic acid. Arachidonic Acid 100-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 34734218-4 2021 Six major xenobiotic metabolizing CYPs were screened for their capacity to catalyze 6:2 FTOH oxidation using chemical inhibitors selective towards CYP isoforms. ftoh 88-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 147-150 34734218-5 2021 Of the CYP isoforms investigated, CYP2A6 was the only enzyme capable of catalyzing 6:2 FTOH in human liver microsomes, with KM and Vmax values of 4076 ng mL-1 and 69 ng mL-1 min-1, respectively. ftoh 87-91 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 7-10 34867397-1 2021 NADPH:cytochrome P450 oxidoreductase (POR) is the obligate electron donor for microsomal cytochrome P450 (CYP) enzymes involved in the biosynthesis of endogenous substances like bile acids and other steroids as well as in the oxidative metabolism of xenobiotics. Bile Acids and Salts 178-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 34867397-1 2021 NADPH:cytochrome P450 oxidoreductase (POR) is the obligate electron donor for microsomal cytochrome P450 (CYP) enzymes involved in the biosynthesis of endogenous substances like bile acids and other steroids as well as in the oxidative metabolism of xenobiotics. Bile Acids and Salts 178-188 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 34867397-1 2021 NADPH:cytochrome P450 oxidoreductase (POR) is the obligate electron donor for microsomal cytochrome P450 (CYP) enzymes involved in the biosynthesis of endogenous substances like bile acids and other steroids as well as in the oxidative metabolism of xenobiotics. Steroids 199-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 34867397-1 2021 NADPH:cytochrome P450 oxidoreductase (POR) is the obligate electron donor for microsomal cytochrome P450 (CYP) enzymes involved in the biosynthesis of endogenous substances like bile acids and other steroids as well as in the oxidative metabolism of xenobiotics. Steroids 199-207 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-109 34328253-3 2021 We aimed to evaluate cytochrome P450 (CYP)-mediated levonorgestrel metabolism and quantify the effects of dolutegravir on levonorgestrel apparent intrinsic clearance (CLint.app. ) Levonorgestrel 52-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 21-36 34328253-3 2021 We aimed to evaluate cytochrome P450 (CYP)-mediated levonorgestrel metabolism and quantify the effects of dolutegravir on levonorgestrel apparent intrinsic clearance (CLint.app. ) Levonorgestrel 52-66 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 38-41 34328253-6 2021 of levonorgestrel was quantified using a recombinant human CYP (rhCYP) enzyme system. Levonorgestrel 3-17 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 59-62 34613323-0 2021 Water biocatalytic effect attenuates cytochrome P450-mediated carcinogenicity of diethylnitrosamine: A computational insight. Water 0-5 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 34613323-0 2021 Water biocatalytic effect attenuates cytochrome P450-mediated carcinogenicity of diethylnitrosamine: A computational insight. Diethylnitrosamine 81-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 34613323-1 2021 The mechanism-based mutagenicity and carcinogenicity of diethylnitrosamine (DEN) are believed to act through interactions with cytochrome P450 (P450) enzymes. Diethylnitrosamine 56-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-142 34613323-1 2021 The mechanism-based mutagenicity and carcinogenicity of diethylnitrosamine (DEN) are believed to act through interactions with cytochrome P450 (P450) enzymes. Diethylnitrosamine 76-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-142 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. noramidopyrine 24-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-172 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. noramidopyrine 24-47 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-177 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. 4-maa 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-172 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. 4-maa 49-54 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-177 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. n-demethylated 64-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-172 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. Ampyrone 82-99 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-172 34663919-3 2021 Here we describe engineered cytochrome P450 enzymes that catalyse carbene N-H insertion to prepare biologically relevant alpha-amino lactones with high activity and enantioselectivity (up to 32,100 total turnovers, >99% yield and 98% e.e.). carbene 66-73 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 34663919-3 2021 Here we describe engineered cytochrome P450 enzymes that catalyse carbene N-H insertion to prepare biologically relevant alpha-amino lactones with high activity and enantioselectivity (up to 32,100 total turnovers, >99% yield and 98% e.e.). alpha-amino lactones 121-141 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 28-43 34848204-4 2022 CYP enzymes can also participate in crosstalk with other PUFA pathways and metabolize prostaglandin G2 and H2, which are the precursors of effector prostaglandins, to affect macrophage function and lymphangiogenesis. prostaglandin G2 86-102 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 34848204-4 2022 CYP enzymes can also participate in crosstalk with other PUFA pathways and metabolize prostaglandin G2 and H2, which are the precursors of effector prostaglandins, to affect macrophage function and lymphangiogenesis. Deuterium 107-109 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 34848204-4 2022 CYP enzymes can also participate in crosstalk with other PUFA pathways and metabolize prostaglandin G2 and H2, which are the precursors of effector prostaglandins, to affect macrophage function and lymphangiogenesis. Prostaglandins 148-162 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-3 34776966-4 2021 Curcumin can inhibit many CYP enzymes in vitro, and so the inhibition of curcumin on CYP enzymes was compared by human liver microsomes, human hepatocytes, and hiHeps using UPLC-MS and the cocktail method. Curcumin 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 26-29 34776966-4 2021 Curcumin can inhibit many CYP enzymes in vitro, and so the inhibition of curcumin on CYP enzymes was compared by human liver microsomes, human hepatocytes, and hiHeps using UPLC-MS and the cocktail method. Curcumin 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-88 34500236-8 2021 Human recombinant cytochrome P450 (CYP) enzyme analysis revealed that metabolism of cabozantinib was mainly catalyzed by CYP3A4, while other CYP enzymes played negligible role. cabozantinib 84-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 18-33 34500236-8 2021 Human recombinant cytochrome P450 (CYP) enzyme analysis revealed that metabolism of cabozantinib was mainly catalyzed by CYP3A4, while other CYP enzymes played negligible role. cabozantinib 84-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 35-38 34672553-5 2021 In this study, we introduce iCYP-MFE, a computational framework for virtual screening on CYP inhibitors toward 1A2, 2C9, 2C19, 2D6, and 3A4 isoforms. icyp-mfe 28-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 34672553-12 2021 An online web server for iCYP-MFE with a user-friendly interface was also deployed to support scientific communities in identifying CYP inhibitors. icyp-mfe 25-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 132-135 34663070-0 2021 Precision Biotransformation of Emerging Pollutants by Human Cytochrome P450 Using Computational-Experimental Synergy: A Case Study of Tris(1,3-dichloro-2-propyl) Phosphate. tris(1,3-dichloro-2-propyl)phosphate 134-171 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 60-75 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. Ampyrone 101-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-172 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. 4-formylaminoantipyrine 122-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-172 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. 4-formylaminoantipyrine 122-145 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-177 34648192-9 2022 CONCLUSION: CYP1A2 is the major CYP for the conversion of 4-MAA to 4-AA and 4-FAA. Ampyrone 67-71 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. 4-formylaminoantipyrine 147-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 157-172 34648192-2 2022 The primary metabolite, 4-methylaminoantipyrine (4-MAA), can be N-demethylated to 4-aminoantipyrine (4-AA) or oxidized to 4-formylaminoantipyrine (4-FAA) by cytochrome P450 (CYP)-dependent reactions. 4-formylaminoantipyrine 147-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 174-177 34648192-9 2022 CONCLUSION: CYP1A2 is the major CYP for the conversion of 4-MAA to 4-AA and 4-FAA. 4-formylaminoantipyrine 76-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 32-35 34648192-3 2022 We aimed to identify the CYPs involved in 4-MAA metabolism and to quantify the effect of CYP inhibition on 4-MAA metabolism. 4-maa 107-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-92 34330718-0 2021 Cytochrome P450-Catalyzed Metabolism of Cannabidiol to the Active Metabolite 7-Hydroxy-Cannabidiol. Cannabidiol 40-51 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34648192-4 2022 METHODS: We investigated the metabolism of 4-MAA in vitro using CYP expressing supersomes and the pharmacokinetics of metamizole in the presence of CYP inhibitors in male subjects. 4-maa 43-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 64-67 34648192-5 2022 RESULTS: The experiments in supersomes revealed CYP1A2 as the major CYP for 4-MAA N-demethylation and 4-FAA formation with CYP2C19 and CYP2D6 contributing to N-demethylation. 4-maa 76-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 34648192-5 2022 RESULTS: The experiments in supersomes revealed CYP1A2 as the major CYP for 4-MAA N-demethylation and 4-FAA formation with CYP2C19 and CYP2D6 contributing to N-demethylation. Nitrogen 82-83 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 34648192-5 2022 RESULTS: The experiments in supersomes revealed CYP1A2 as the major CYP for 4-MAA N-demethylation and 4-FAA formation with CYP2C19 and CYP2D6 contributing to N-demethylation. 4-formylaminoantipyrine 102-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 68-71 34690761-0 2021 Effects of Cytochrome P450 and Transporter Polymorphisms on the Bioavailability and Safety of Dutasteride and Tamsulosin. Dutasteride 94-105 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-26 34690761-0 2021 Effects of Cytochrome P450 and Transporter Polymorphisms on the Bioavailability and Safety of Dutasteride and Tamsulosin. Tamsulosin 110-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-26 34330718-0 2021 Cytochrome P450-Catalyzed Metabolism of Cannabidiol to the Active Metabolite 7-Hydroxy-Cannabidiol. 7-Hydroxycannabidiol 77-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34620272-3 2021 Cytochrome-P450 (CYP) isoenzymes CYP3A4 and CYP3A5, as well P-glycoprotein (P-gp) are involved in TAC bioavailability. Tacrolimus 98-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34620272-3 2021 Cytochrome-P450 (CYP) isoenzymes CYP3A4 and CYP3A5, as well P-glycoprotein (P-gp) are involved in TAC bioavailability. Tacrolimus 98-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 17-20 34180113-0 2021 Enzymatic Tailoring in Luzopeptin Biosynthesis Involves Cytochrome P450-Mediated Carbon-Nitrogen Bond Desaturation for Hydrazone Formation. luzopeptin 23-33 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 34556703-8 2021 Significantly, our findings suggest a potential targeted therapeutic approach to manage HNCs by exploiting intratumoural CYP expression for metabolic activation of duocarmycin-based prodrugs such as ICT2700. (1-(chloromethyl)-1,2-dihydropyrrolo(3,2-e)indol-3(6H)-yl)(5-methoxy-1H-indol-2-yl)methanone 199-206 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 121-124 34563259-5 2021 Studies with ovarian theca cells taken from PCOS women have demonstrated increased androgen production due to augmented ovarian steroidogenesis attributed to mainly altered expression of critical enzymes (Cytochrome P450 enzymes: CYP17, CYP21, CYP19, CYP11A) in the steroid hormone biosynthesis pathway. Steroids 266-273 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 205-220 34180113-0 2021 Enzymatic Tailoring in Luzopeptin Biosynthesis Involves Cytochrome P450-Mediated Carbon-Nitrogen Bond Desaturation for Hydrazone Formation. Carbon 81-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 34180113-0 2021 Enzymatic Tailoring in Luzopeptin Biosynthesis Involves Cytochrome P450-Mediated Carbon-Nitrogen Bond Desaturation for Hydrazone Formation. Nitrogen 88-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 34180113-0 2021 Enzymatic Tailoring in Luzopeptin Biosynthesis Involves Cytochrome P450-Mediated Carbon-Nitrogen Bond Desaturation for Hydrazone Formation. Hydrazones 119-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 56-71 34180113-3 2021 Significantly, we revealed a multitasking cytochrome P450 enzyme that catalyzes four consecutive oxidations including the highly unusual carbon-nitrogen bond desaturation, forming the hydrazone-bearing 4-OH-Thp residues. Carbon 137-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 34180113-3 2021 Significantly, we revealed a multitasking cytochrome P450 enzyme that catalyzes four consecutive oxidations including the highly unusual carbon-nitrogen bond desaturation, forming the hydrazone-bearing 4-OH-Thp residues. Nitrogen 144-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 34180113-3 2021 Significantly, we revealed a multitasking cytochrome P450 enzyme that catalyzes four consecutive oxidations including the highly unusual carbon-nitrogen bond desaturation, forming the hydrazone-bearing 4-OH-Thp residues. Hydrazones 184-193 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 34180113-3 2021 Significantly, we revealed a multitasking cytochrome P450 enzyme that catalyzes four consecutive oxidations including the highly unusual carbon-nitrogen bond desaturation, forming the hydrazone-bearing 4-OH-Thp residues. 4-oh-thp 202-210 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 42-57 34114066-1 2021 PURPOSE: Genetic variation in the activation of the prodrug cyclophosphamide (CP) by cytochrome P450 (CYP) enzymes has been shown to influence outcomes. Cyclophosphamide 60-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 34114066-1 2021 PURPOSE: Genetic variation in the activation of the prodrug cyclophosphamide (CP) by cytochrome P450 (CYP) enzymes has been shown to influence outcomes. Cyclophosphamide 60-76 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 102-105 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). Tryptophan 108-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 34330191-0 2021 Evaluation of species differences in the metabolism of the selective NaV1.7 inhibitor DS-1971a, a mixed substrate of cytochrome P450 and aldehyde oxidase. DS-1971a 86-94 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 117-153 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). Tryptophan 108-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). Nitric Oxide 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). Tryptophan 122-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). Nitric Oxide 60-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). Tryptophan 122-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). 4-nitro-l-tryptophan 135-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). 4-nitro-l-tryptophan 135-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). 4-no2-trp 157-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-25 34319079-1 2021 TxtE is a cytochrome P450 (CYP) homologue that mediates the nitric oxide (NO)-dependent direct nitration of l-tryptophan (Trp) to form 4-nitro-l-tryptophan (4-NO2-Trp). 4-no2-trp 157-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-30 34170130-5 2021 PCZ-treated cells activated intracellular oxidative stress via cytochrome P450 and had higher mRNA levels of interleukin-1beta, tumor necrosis factor-alpha, matrix metalloproteinase (MMP)-2, MMP-9, and transforming growth factor-beta (TGF-beta) than the control. propiconazole 0-3 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-78 34273352-1 2021 Cytochrome P450 (P450) 17A1 catalyzes the 17alpha-hydroxylation of progesterone and pregnenolone as well as the subsequent lyase cleavage of both products to generate androgens. Progesterone 67-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-27 34273352-1 2021 Cytochrome P450 (P450) 17A1 catalyzes the 17alpha-hydroxylation of progesterone and pregnenolone as well as the subsequent lyase cleavage of both products to generate androgens. Pregnenolone 84-96 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-27 34138544-0 2021 Correction to Bifunctional Cytochrome P450 Enzymes Involved in Camptothecin Biosynthesis. Camptothecin 63-75 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 27-42 34101429-0 2021 Cytochrome P450-Mediated Metabolism and CYP Inhibition for the Synthetic Peroxide Antimalarial OZ439. Peroxides 73-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34101429-0 2021 Cytochrome P450-Mediated Metabolism and CYP Inhibition for the Synthetic Peroxide Antimalarial OZ439. artefenomel 95-100 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34143391-5 2021 Lenabasum is a CYP substrate, and the model predicted lenabasum clearance of 51% by CYP2C9, 37% by CYP2C8, and 12% by CYP3A4. lenabasum 0-9 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 34143391-5 2021 Lenabasum is a CYP substrate, and the model predicted lenabasum clearance of 51% by CYP2C9, 37% by CYP2C8, and 12% by CYP3A4. lenabasum 54-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 15-18 34143391-8 2021 The simulations of CYP inducers predicted that the strongest interaction would occur with rifampin, with the AUC decreasing to 0.36 of the control value, whereas the simulations of CYP inhibitors predicted that the greatest effect would occur with fluconazole, with a 1.43-fold increase in AUC. Rifampin 90-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 19-22 34143391-8 2021 The simulations of CYP inducers predicted that the strongest interaction would occur with rifampin, with the AUC decreasing to 0.36 of the control value, whereas the simulations of CYP inhibitors predicted that the greatest effect would occur with fluconazole, with a 1.43-fold increase in AUC. Fluconazole 248-259 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 181-184 34327042-1 2021 Bacterial cytochrome P450 (P450) 101A1 (P450cam) has served as a prototype among the P450 enzymes and has high catalytic activity towards its cognate substrate, camphor. Camphor 161-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 10-38 34120382-0 2021 Targeted analysis of eicosanoids derived from cytochrome P450 pathway by high-resolution multiple-reaction monitoring mass spectrometry. Eicosanoids 21-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-61 34120382-1 2021 Cytochrome P450 (CYP450) pathway is one of the critical enzymatic via eicosanoid biosynthesis. Eicosanoids 70-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 34113252-0 2021 Interactive Effects of Glucocorticoids and Cytochrome P450 Polymorphisms on the Plasma Trough Concentrations of Voriconazole. Voriconazole 112-124 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 43-58 34113252-1 2021 Aims: To explore the interactive influence of glucocorticoids and cytochrome P450 (CYP450) polymorphisms on voriconazole (VRC) plasma trough concentrations (Cmin) and provide a reliable basis for reasonable application of VRC. Voriconazole 108-120 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 34113252-1 2021 Aims: To explore the interactive influence of glucocorticoids and cytochrome P450 (CYP450) polymorphisms on voriconazole (VRC) plasma trough concentrations (Cmin) and provide a reliable basis for reasonable application of VRC. Voriconazole 122-125 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 34163706-4 2021 The reaction of 2 with a triphenylcarbon radical further gives triphenylmethanol and mimics the so-called oxygen rebound step of Cpd II of cytochrome P450. triphenylcarbon radical 25-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 34163706-4 2021 The reaction of 2 with a triphenylcarbon radical further gives triphenylmethanol and mimics the so-called oxygen rebound step of Cpd II of cytochrome P450. TRIPHENYLMETHANOL 63-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 34163706-4 2021 The reaction of 2 with a triphenylcarbon radical further gives triphenylmethanol and mimics the so-called oxygen rebound step of Cpd II of cytochrome P450. Oxygen 106-112 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 139-154 34334558-7 2021 In the presence of human liver cytosolic fractions, the oxidative formation rate of o-hydroxyphenylacetic acid was relatively high in cytochrome P450 (P450) 1A2-rich human liver microsomes. 2-hydroxyphenylacetic acid 84-110 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 134-149 35611779-6 2022 The pharmacokinetics of many antiseizure medications is not influenced by antipsychotics and anxiolytics while some newer antidepressants namely fluoxetine, fluvoxamine and viloxazine, may inhibit CYP enzymes leading to increased serum concentrations of some antiseizure medications including phenytoin and carbamazepine. Fluoxetine 145-155 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-200 34272716-8 2021 Simultaneously docking of (S)-warfarin and ABT revealed that (S)-warfarin outcompetes ABT for the distal binding site and results in the binding of ABT to the CYP2C9 active site, supporting the observations of potent inactivation of CYP2C9 when (S)-warfarin is the probe substrate.These results highlight that probe selection is crucial when evaluating CYP inhibition potential, and it is recommended that multiple probes be utilized for CYP2C9, similar to the approach routinely employed for CYP3A4. 1-aminobenzotriazole 148-151 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 353-356 34272716-9 2021 Further, utilization of ABT as a pan-inhibitor of CYP activity for investigational compounds, both in vitro and in vivo, should be accompanied with the understanding that residual CYP-mediated oxidative metabolism could potentially be observed for CYP2C9 substrates and should not necessarily be attributed to non-P450-mediated metabolism. 1-aminobenzotriazole 24-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 50-53 34272716-9 2021 Further, utilization of ABT as a pan-inhibitor of CYP activity for investigational compounds, both in vitro and in vivo, should be accompanied with the understanding that residual CYP-mediated oxidative metabolism could potentially be observed for CYP2C9 substrates and should not necessarily be attributed to non-P450-mediated metabolism. 1-aminobenzotriazole 24-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 34272718-4 2021 Specifically, a case is presented in which reduced glutathione (GSH) was included in an experiment with human liver S9 fraction to trap reactive metabolites generated from cytochrome P450-mediated metabolism of lapatinib and its O-dealkylated metabolite, M1 (question 1). Glutathione 51-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-187 34272718-4 2021 Specifically, a case is presented in which reduced glutathione (GSH) was included in an experiment with human liver S9 fraction to trap reactive metabolites generated from cytochrome P450-mediated metabolism of lapatinib and its O-dealkylated metabolite, M1 (question 1). Glutathione 64-67 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-187 34272718-4 2021 Specifically, a case is presented in which reduced glutathione (GSH) was included in an experiment with human liver S9 fraction to trap reactive metabolites generated from cytochrome P450-mediated metabolism of lapatinib and its O-dealkylated metabolite, M1 (question 1). Lapatinib 211-220 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 172-187 35523303-3 2022 To clarify the role of cholesterol (Chol) in the CYP-related drug metabolic process, we investigated the lipid bilayer entry of CYP-substrate drugs using a model membrane system as follows. Lipid Bilayers 105-118 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 128-131 35523303-6 2022 Chlorzoxazone (CZX) was used as the CYP-substrate drug. Chlorzoxazone 0-13 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 35523303-6 2022 Chlorzoxazone (CZX) was used as the CYP-substrate drug. Chlorzoxazone 15-18 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 36-39 35483145-0 2022 In Silico simulation of Cytochrome P450-Mediated metabolism of aromatic amines: A case study of N-Hydroxylation. aromatic amines 63-78 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 35483145-0 2022 In Silico simulation of Cytochrome P450-Mediated metabolism of aromatic amines: A case study of N-Hydroxylation. Nitrogen 96-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). Diclofenac 40-50 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 442-445 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). 1-aminobenzotriazole 458-478 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 442-445 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). 1-aminobenzotriazole 480-483 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 442-445 35523303-0 2022 Cholesterol"s inhibition effect on entering of chlorzoxazone into phosphatidylethanolamine bilayer: Relevance to cytochrome P450 drug metabolism at endoplasmic reticulum membranes. Cholesterol 0-11 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 35523303-0 2022 Cholesterol"s inhibition effect on entering of chlorzoxazone into phosphatidylethanolamine bilayer: Relevance to cytochrome P450 drug metabolism at endoplasmic reticulum membranes. Chlorzoxazone 47-60 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 35523303-0 2022 Cholesterol"s inhibition effect on entering of chlorzoxazone into phosphatidylethanolamine bilayer: Relevance to cytochrome P450 drug metabolism at endoplasmic reticulum membranes. phosphatidylethanolamine bilayer 66-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 113-128 35594715-0 2022 Interactions of resveratrol and its metabolites (resveratrol-3-sulfate, resveratrol-3-glucuronide, and dihydroresveratrol) with serum albumin, cytochrome P450 enzymes, and OATP transporters. Resveratrol 16-27 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 143-158 35594715-4 2022 Therefore, in the current study, we examined the interactions of resveratrol-3-sulfate (R3S), resveratrol-3-glucuronide, and dihydroresveratrol (DHR; a metabolite produced by the colon microbiota) with human serum albumin (HSA), cytochrome P450 (CYP) enzymes, and organic anion transporting polypeptides (OATP) employing in vitro models. resveratrol-3-glucuronide 94-119 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 229-244 35594715-4 2022 Therefore, in the current study, we examined the interactions of resveratrol-3-sulfate (R3S), resveratrol-3-glucuronide, and dihydroresveratrol (DHR; a metabolite produced by the colon microbiota) with human serum albumin (HSA), cytochrome P450 (CYP) enzymes, and organic anion transporting polypeptides (OATP) employing in vitro models. dihydroresveratrol 125-143 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 229-244 35611779-6 2022 The pharmacokinetics of many antiseizure medications is not influenced by antipsychotics and anxiolytics while some newer antidepressants namely fluoxetine, fluvoxamine and viloxazine, may inhibit CYP enzymes leading to increased serum concentrations of some antiseizure medications including phenytoin and carbamazepine. Fluvoxamine 157-168 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-200 35611779-6 2022 The pharmacokinetics of many antiseizure medications is not influenced by antipsychotics and anxiolytics while some newer antidepressants namely fluoxetine, fluvoxamine and viloxazine, may inhibit CYP enzymes leading to increased serum concentrations of some antiseizure medications including phenytoin and carbamazepine. Viloxazine 173-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-200 35611779-6 2022 The pharmacokinetics of many antiseizure medications is not influenced by antipsychotics and anxiolytics while some newer antidepressants namely fluoxetine, fluvoxamine and viloxazine, may inhibit CYP enzymes leading to increased serum concentrations of some antiseizure medications including phenytoin and carbamazepine. Phenytoin 293-302 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-200 35611779-6 2022 The pharmacokinetics of many antiseizure medications is not influenced by antipsychotics and anxiolytics while some newer antidepressants namely fluoxetine, fluvoxamine and viloxazine, may inhibit CYP enzymes leading to increased serum concentrations of some antiseizure medications including phenytoin and carbamazepine. Carbamazepine 307-320 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 197-200 35562949-3 2022 The electrophiles resulting from the cytochrome P450-mediated metabolism of N-nitrosamines can readily react with DNA and form covalent addition products (DNA adducts) that play a central role in carcinogenesis if not repaired. n-nitrosamines 76-90 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 37-52 35527244-0 2022 Influence of cytochrome P450 and glutathione S transferase polymorphisms on response to nilotinib therapy among chronic myeloidleukemia patients from Pakistan. nilotinib 88-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 13-28 35578608-16 2022 The metabolite data demonstrated increasing CYP metabolites in occurrence with the delayed acetaminophen peak concentration. Acetaminophen 91-104 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 44-47 35289081-0 2022 Application of physiologically based pharmacokinetic modelling for the prediction of drug-drug interactions involving anlotinib as a perpetrator of cytochrome P450 enzymes. anlotinib 118-127 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 148-163 35289081-3 2022 In vitro experiments demonstrated that anlotinib is a substrate of cytochrome P450 (CYP) enzymes and moderate inhibitor of several common ones; however, no clinical DDI studies have been performed to investigate inhibitory effects of anlotinib on these CYP enzymes. anlotinib 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 67-82 35289081-3 2022 In vitro experiments demonstrated that anlotinib is a substrate of cytochrome P450 (CYP) enzymes and moderate inhibitor of several common ones; however, no clinical DDI studies have been performed to investigate inhibitory effects of anlotinib on these CYP enzymes. anlotinib 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 84-87 35289081-3 2022 In vitro experiments demonstrated that anlotinib is a substrate of cytochrome P450 (CYP) enzymes and moderate inhibitor of several common ones; however, no clinical DDI studies have been performed to investigate inhibitory effects of anlotinib on these CYP enzymes. anlotinib 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 253-256 35289081-3 2022 In vitro experiments demonstrated that anlotinib is a substrate of cytochrome P450 (CYP) enzymes and moderate inhibitor of several common ones; however, no clinical DDI studies have been performed to investigate inhibitory effects of anlotinib on these CYP enzymes. anlotinib 234-243 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 253-256 35289081-7 2022 As a result, the marginal plasma exposure changes of typical CYP3A and CYP2C9 substrates were less than the bioequivalence threshold, indicating that anlotinib has a very low potential of causing clinically meaningful DDI through the inhibition of several major CYP enzymes. anlotinib 150-159 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 262-265 35115300-0 2022 Comprehensive predictions of cytochrome P450 (CYP)-mediated in vivo cannabinoid-drug interactions based on reversible and time-dependent CYP inhibition in human liver microsomes. Cannabinoids 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 29-44 35302484-2 2022 In the drug combination nirmatrelvir + ritonavir (nirmatrelvir/r), the active agent, nirmatrelvir, is made bioavailable in clinically adequate amounts by the additional administration of a potent inhibitor of its first-pass metabolism by way of cytochrome P450 (CYP) 3A in the gut and liver. 7R9A5P7H32 24-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 245-269 35302484-2 2022 In the drug combination nirmatrelvir + ritonavir (nirmatrelvir/r), the active agent, nirmatrelvir, is made bioavailable in clinically adequate amounts by the additional administration of a potent inhibitor of its first-pass metabolism by way of cytochrome P450 (CYP) 3A in the gut and liver. Ritonavir 39-48 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 245-269 35302484-2 2022 In the drug combination nirmatrelvir + ritonavir (nirmatrelvir/r), the active agent, nirmatrelvir, is made bioavailable in clinically adequate amounts by the additional administration of a potent inhibitor of its first-pass metabolism by way of cytochrome P450 (CYP) 3A in the gut and liver. 7R9A5P7H32 50-62 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 245-269 35302484-2 2022 In the drug combination nirmatrelvir + ritonavir (nirmatrelvir/r), the active agent, nirmatrelvir, is made bioavailable in clinically adequate amounts by the additional administration of a potent inhibitor of its first-pass metabolism by way of cytochrome P450 (CYP) 3A in the gut and liver. Arginine 63-64 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 245-269 35302484-2 2022 In the drug combination nirmatrelvir + ritonavir (nirmatrelvir/r), the active agent, nirmatrelvir, is made bioavailable in clinically adequate amounts by the additional administration of a potent inhibitor of its first-pass metabolism by way of cytochrome P450 (CYP) 3A in the gut and liver. 7R9A5P7H32 85-97 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 245-269 35348513-1 2022 Budesonide (Tarpeyo) has received accelerated approval to reduce proteinuria in adults with primary immunoglobulin A nephropathy, also known as Berger"s disease.Nurses should assess if any coprescribed drugs could induce a drug-drug interaction via the cytochrome P-450 isoenzyme system. Budesonide 0-10 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 253-269 35348513-1 2022 Budesonide (Tarpeyo) has received accelerated approval to reduce proteinuria in adults with primary immunoglobulin A nephropathy, also known as Berger"s disease.Nurses should assess if any coprescribed drugs could induce a drug-drug interaction via the cytochrome P-450 isoenzyme system. tarpeyo 12-19 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 253-269 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Dronabinol 164-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-238 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Dronabinol 164-167 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-243 35115300-10 2022 Significance Statement This study, combined with our previous findings, provides for the first time a comprehensive analysis of the potential for cannabidiol, delta-9-tetrahydrocannabinol, and their metabolites to inhibit cytochrome P450 enzymes in a reversible or time-dependent manner. Cannabidiol 146-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-237 35115300-10 2022 Significance Statement This study, combined with our previous findings, provides for the first time a comprehensive analysis of the potential for cannabidiol, delta-9-tetrahydrocannabinol, and their metabolites to inhibit cytochrome P450 enzymes in a reversible or time-dependent manner. Dronabinol 159-187 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 222-237 35115300-0 2022 Comprehensive predictions of cytochrome P450 (CYP)-mediated in vivo cannabinoid-drug interactions based on reversible and time-dependent CYP inhibition in human liver microsomes. Cannabinoids 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 46-49 35115300-0 2022 Comprehensive predictions of cytochrome P450 (CYP)-mediated in vivo cannabinoid-drug interactions based on reversible and time-dependent CYP inhibition in human liver microsomes. Cannabinoids 68-79 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 137-140 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Cannabidiol 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-238 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Cannabidiol 105-116 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-243 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Cannabidiol 118-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-238 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Cannabidiol 118-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-243 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Dronabinol 124-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-238 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Dronabinol 124-152 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-243 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Dronabinol 154-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 223-238 35115300-1 2022 We previously reported the unbound reversible (IC50,u) and time-dependent (KI,u) inhibition potencies of cannabidiol (CBD), delta-9-tetrahydrocannabinol (THC), and THC metabolites (11-OH THC, 11-COOH THC) against the major cytochrome P450 (CYP) enzymes (1A2, 2C9, 2C19, 2D6, 3A). Dronabinol 154-157 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 240-243 35431771-0 2022 Methylated Cytochrome P450 and the Solute Carrier Family of Genes Correlate With Perturbations in Bile Acid Metabolism in Parkinson"s Disease. Bile Acids and Salts 98-107 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 11-26 35408976-0 2022 Molecular Lego of Human Cytochrome P450: The Key Role of Heme Domain Flexibility for the Activity of the Chimeric Proteins. Heme 57-61 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 24-39 35408976-1 2022 The cytochrome P450 superfamily are heme-thiolate enzymes able to carry out monooxygenase reactions. Heme 36-40 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 35408976-1 2022 The cytochrome P450 superfamily are heme-thiolate enzymes able to carry out monooxygenase reactions. thiolate 41-49 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-19 35230092-0 2022 Computational Investigation of the Bisphenolic Drug Metabolism by Cytochrome P450: What Factors Favor Intramolecular Phenol Coupling. Phenol 117-123 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 35230092-1 2022 Intramolecular phenol coupling reactions of alkaloids can lead to active metabolites catalyzed by the mammalian cytochrome P450 enzyme (P450); however, the mechanistic knowledge of such an "unusual" process is lacking. Phenol 15-21 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-127 35230092-1 2022 Intramolecular phenol coupling reactions of alkaloids can lead to active metabolites catalyzed by the mammalian cytochrome P450 enzyme (P450); however, the mechanistic knowledge of such an "unusual" process is lacking. Alkaloids 44-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 112-127 35121066-0 2022 The impact of legacy and novel perfluoroalkyl substances on human cytochrome P450: An in vitro study on the inhibitory potential and underlying mechanisms. perfluoroalkyl substances 31-56 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 66-81 35386678-9 2022 Pathway enrichment analysis shows that "phenylpropanoid biosynthesis"/"cutin, suberin, and wax biosynthesis" is significantly enriched in these groups, while the AC vs. AC and PVP groups are mainly enriched in "cytochrome P450," indicating that AC may promote the further development of roots into seedlings by stimulating "phenylpropanoid biosynthesis" and biosynthesis of stratum cutin and suberin. phenylpropanoid 40-55 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 211-226 35107462-6 2022 Finally, the metabolite identification opportunities were also explored using efavirenz as an example drug with complex primary and secondary metabolism involving a combination of CYP, UGT and sulfotransferase enzymes. efavirenz 78-87 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 35386280-12 2022 GSEA revealed samples with MDN1 mutation enriched in retinol metabolism, drug metabolism cytochrome P450, glucuronidation, miscellaneous transport, and binding event pathways. gsea 0-4 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 89-104 35617891-2 2022 In this study, we explored the possibility of using prokaryotic water-soluble cytochrome P450 (CYP) to produce human metabolites. Water 64-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 78-93 35617891-2 2022 In this study, we explored the possibility of using prokaryotic water-soluble cytochrome P450 (CYP) to produce human metabolites. Water 64-69 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 95-98 35121066-3 2022 Recently, the interaction with cytochrome P450 (CYP) has been proposed as a potential key molecular event leading to PFAS-induced hepatotoxicity. pfas 117-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 31-46 35121066-3 2022 Recently, the interaction with cytochrome P450 (CYP) has been proposed as a potential key molecular event leading to PFAS-induced hepatotoxicity. pfas 117-121 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 48-51 35121066-6 2022 Using the same assays, Michaelis-Menten saturation curves were determined to explore the type of PFAS-induced CYP inhibition. pfas 97-101 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 110-113 35194103-1 2022 Human CYP2B6 enzyme although constitutes relatively low proportion (1-4%) of hepatic cytochrome P450 content, it is the major catalyst of metabolism of several clinically important drugs (efavirenz, cyclophosphamide, bupropion, methadone). efavirenz 188-197 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 35280254-2 2021 Caffeine is almost exclusively metabolized in the liver by the cytochrome P-450 enzyme system to the main product paraxanthine and the additional products theobromine and theophylline. Caffeine 0-8 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 35280254-2 2021 Caffeine is almost exclusively metabolized in the liver by the cytochrome P-450 enzyme system to the main product paraxanthine and the additional products theobromine and theophylline. 1,7-dimethylxanthine 114-126 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 35280254-2 2021 Caffeine is almost exclusively metabolized in the liver by the cytochrome P-450 enzyme system to the main product paraxanthine and the additional products theobromine and theophylline. Theobromine 155-166 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 35280254-2 2021 Caffeine is almost exclusively metabolized in the liver by the cytochrome P-450 enzyme system to the main product paraxanthine and the additional products theobromine and theophylline. Theophylline 171-183 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 63-79 35600548-0 2022 Cytochrome P450 enzymes mediated by DNA methylation is involved in deoxynivalenol-induced hepatoxicity in piglets. deoxynivalenol 67-81 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 35194103-1 2022 Human CYP2B6 enzyme although constitutes relatively low proportion (1-4%) of hepatic cytochrome P450 content, it is the major catalyst of metabolism of several clinically important drugs (efavirenz, cyclophosphamide, bupropion, methadone). Cyclophosphamide 199-215 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 35194103-1 2022 Human CYP2B6 enzyme although constitutes relatively low proportion (1-4%) of hepatic cytochrome P450 content, it is the major catalyst of metabolism of several clinically important drugs (efavirenz, cyclophosphamide, bupropion, methadone). Bupropion 217-226 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 35194103-1 2022 Human CYP2B6 enzyme although constitutes relatively low proportion (1-4%) of hepatic cytochrome P450 content, it is the major catalyst of metabolism of several clinically important drugs (efavirenz, cyclophosphamide, bupropion, methadone). Methadone 228-237 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 85-100 35180866-0 2022 In vitro evaluation of the inhibition potential of echinacoside on human cytochrome P450 isozymes. echinacoside 51-63 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 73-88 35135554-0 2022 Relationship of cytochrome P450 gene polymorphisms with blood concentrations of hydroxychloroquine and its metabolites and adverse drug reactions. Hydroxychloroquine 80-98 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 16-31 35185404-7 2022 In elderly or frail individuals who are on multiple comedications (particularly if >=1 moderate or strong cytochrome P-450 inhibitor), only edoxaban consistently reduced major bleeding compared to warfarin. edoxaban 140-148 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 106-122 35135554-2 2022 This study aimed to investigate the relationship of cytochrome P450 (CYP450) gene polymorphisms with blood concentrations of HCQ and its metabolites and adverse drug reactions (ADRs) in patients with SLE and RA. Hydroxychloroquine 125-128 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 52-67 33543792-8 2022 Using FAERS data, only those DDI-ADE associations were considered that met the disproportionality threshold of Omega > 0 for a CYP substrate when paired with at least two inhibitors. ddi-ade 29-36 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 127-130 35132941-5 2022 To investigate the activation pathway of synthesized N-formyl pyrazoline substituted steroid derivatives, a molecular docking study was performed on human cytochrome P450-(CYP17A1: PDB ID 5IRQ) with the help of the free AutoDock Vina. n-formyl pyrazoline 53-72 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 35132941-5 2022 To investigate the activation pathway of synthesized N-formyl pyrazoline substituted steroid derivatives, a molecular docking study was performed on human cytochrome P450-(CYP17A1: PDB ID 5IRQ) with the help of the free AutoDock Vina. Steroids 85-92 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 155-170 34380995-3 2022 The mechanism of methadone metabolism and disposition has been shown to involve cytochrome P450 (CYP450) and P-glycoprotein. Methadone 17-26 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 80-95 34996326-6 2022 The other important clinical applications of piperine are due to its bio enhancing effect on drugs, by modulating, absorption in the gastrointestinal tract, altering activities of transporters like p-glycoprotein substrates, and modulating drug metabolism by altering the expression of cytochrome P450 or UDP-glucuronosyltransferase enzymes. piperine 45-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 286-301