PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 31324697-0 2019 Efavirenz Metabolism: Influence of Polymorphic CYP2B6 Variants and Stereochemistry. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 31306645-6 2019 In addition, some CAR antagonists which repress CYP2B6 mRNA expression in HPH, such as sorafenib, rimonabant, and CINPA1, were found to translocate hCAR to the nucleus of HPH. Sorafenib 87-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-54 31006110-2 2019 Efavirenz is predominantly metabolized into inactive metabolites by cytochrome P450 (CYP)2B6, and patients with certain CYP2B6 genetic variants may be at increased risk for adverse effects, particularly central nervous system toxicity and treatment discontinuation. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-92 31006110-2 2019 Efavirenz is predominantly metabolized into inactive metabolites by cytochrome P450 (CYP)2B6, and patients with certain CYP2B6 genetic variants may be at increased risk for adverse effects, particularly central nervous system toxicity and treatment discontinuation. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 31006110-3 2019 We summarize the evidence from the literature and provide therapeutic recommendations for efavirenz prescribing based on CYP2B6 genotypes. efavirenz 90-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-127 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-3 2019 Cytochrome P450 2B6 (CYP2B6) is the major enzyme catalyzing S-8-hydroxyefavirenz formation. 8-hydroxyefavirenz 60-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 31324697-3 2019 Cytochrome P450 2B6 (CYP2B6) is the major enzyme catalyzing S-8-hydroxyefavirenz formation. 8-hydroxyefavirenz 60-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 31324697-4 2019 CYP2B6 genetics and drug interactions are major determinants of clinical efavirenz disposition and dose adjustment. efavirenz 73-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 31324697-5 2019 In addition, as a prototypic CYP2B6 substrate, S-efavirenz and analogs can inform on the structure, activity, catalytic mechanisms, and stereoselectivity of CYP2B6. sefavirenz 47-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 31324697-5 2019 In addition, as a prototypic CYP2B6 substrate, S-efavirenz and analogs can inform on the structure, activity, catalytic mechanisms, and stereoselectivity of CYP2B6. sefavirenz 47-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 31324697-6 2019 Metabolism of R-efavirenz by CYP2B6 remains unexplored. efavirenz 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 31324697-7 2019 This investigation assessed S-efavirenz metabolism by clinically relevant CYP2B6 genetic variants. sefavirenz 28-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 31324697-8 2019 This investigation also evaluated R-efavirenz hydroxylation by wild-type CYP2B6.1 and CYP2B6 variants. efavirenz 34-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 31324697-9 2019 S-Efavirenz 8-hydroxylation by wild-type CYP2B6.1 and variants exhibited positive cooperativity and apparent cooperative substrate inhibition. sefavirenz 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-10 2019 On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 CYP2B6.5 CYP2B6.17 > CYP2B6.6 CYP2B6.7 CYP2B6.9 CYP2B6.19 CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. sefavirenz 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 31324697-11 2019 Rates of R-efavirenz metabolism were approximately 1/10 those of S-efavirenz for wild-type CYP2B6.1 and variants. efavirenz 9-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 31324697-11 2019 Rates of R-efavirenz metabolism were approximately 1/10 those of S-efavirenz for wild-type CYP2B6.1 and variants. sefavirenz 65-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 31324697-12 2019 On the basis of Clmax values, there was 14-fold enantioselectivity (S > R-efavirenz) for wild-type CYP2B6.1, and 5- to 22-fold differences for other CYP2B6 variants. efavirenz 72-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 30825074-4 2019 METHODS: This study was performed to investigate the inhibitory effects of myricetin on human CYP3A4, CYP2B6, CYP2C9, CYP2D6 and rat CYP3A2, CYP2B1, CYP2C11, CYP2D1 through the cocktail approach using ultra-performance liquid chromatography tandem mass spectrometry. myricetin 75-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 30993551-1 2019 BACKGROUND AND OBJECTIVES: A recent report indicated that the pharmacodynamic interaction between clopidogrel and vonoprazan leading to attenuation of the anti-platelet effect of clopidogrel was unlikely to be caused by the inhibition of cytochrome P450 (CYP) 2B6, CYP2C19, or CYP3A4/5 by vonoprazan, based on in vitro CYP inhibition data. Clopidogrel 98-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 238-263 30993551-1 2019 BACKGROUND AND OBJECTIVES: A recent report indicated that the pharmacodynamic interaction between clopidogrel and vonoprazan leading to attenuation of the anti-platelet effect of clopidogrel was unlikely to be caused by the inhibition of cytochrome P450 (CYP) 2B6, CYP2C19, or CYP3A4/5 by vonoprazan, based on in vitro CYP inhibition data. 1-(5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine 114-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 238-263 30993551-2 2019 The current report investigates another important antiplatelet inhibitor, prasugrel, that is also activated through metabolism by CYP2B6, CYP2C19 and CYP3A4/5, for its CYP-based DDI potential with vonoprazan. Prasugrel Hydrochloride 74-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 30993551-2 2019 The current report investigates another important antiplatelet inhibitor, prasugrel, that is also activated through metabolism by CYP2B6, CYP2C19 and CYP3A4/5, for its CYP-based DDI potential with vonoprazan. Didanosine 178-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 30993551-2 2019 The current report investigates another important antiplatelet inhibitor, prasugrel, that is also activated through metabolism by CYP2B6, CYP2C19 and CYP3A4/5, for its CYP-based DDI potential with vonoprazan. 1-(5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine 197-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 31375472-2 2019 The assay was created to develop physiologically relevant P450 inhibition information, taking advantage of the complete organelle composition and their associated drug-metabolizing enzymes of the MMHH but with the ease of use of human liver microsomes, including storage at -80 C instead of in liquid nitrogen, and thaw and use without centrifugation and microscopic evaluation as required for intact hepatocytes. Nitrogen 301-309 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. Sulfur 184-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 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 2 subfamily B member 6 Homo sapiens 127-131 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. Sulfur 184-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. Sulfur 184-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. Sulfur 184-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. Sulfur 184-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 186-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 186-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 186-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 186-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 186-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 291-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 291-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 291-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 291-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-13 2019 These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. efavirenz 291-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31324697-14 2019 Efavirenz is the most stereoselective CYP2B6 drug substrate yet identified and may be a useful probe for the CYP2B6 active site and catalytic mechanisms. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 31324697-14 2019 Efavirenz is the most stereoselective CYP2B6 drug substrate yet identified and may be a useful probe for the CYP2B6 active site and catalytic mechanisms. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 31324697-15 2019 SIGNIFICANCE STATEMENT: Clinical disposition of the antiretroviral S-efavirenz is affected by CYP2B6 polymorphisms. sefavirenz 67-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 31247375-2 2019 As a prodrug, CPA is activated by cytochrome P450 2B6 (CYP2B6), which is transcriptionally regulated by the human constitutive androstane receptor (hCAR). Cyclophosphamide 14-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-53 31324697-16 2019 Expressed CYP2B6 with 516G>T (CYP2B6*6 and CYP2B6*9), and 983T>C (CYP2B6*16 and CYP2B6*18) polymorphisms had a diminishment or loss of function for efavirenz 8-hydroxylation. efavirenz 148-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 31247375-2 2019 As a prodrug, CPA is activated by cytochrome P450 2B6 (CYP2B6), which is transcriptionally regulated by the human constitutive androstane receptor (hCAR). Cyclophosphamide 14-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 31324697-19 2019 With greater than 10-fold enantioselectivity (S- vs. R- metabolism), efavirenz is the most stereoselective CYP2B6 drug substrate yet identified. efavirenz 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 31270214-5 2019 In addition, the knockdown of ADAR1 resulted in the enhanced induction of CYP2B6 and CYP3A4 mRNA by 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime and phenobarbital, respectively. 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde 100-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 31247375-6 2019 DL5016 robustly induced the expression of hCAR target gene CYP2B6, at both the mRNA and protein levels, and caused translocation of hCAR from the cytoplasm to the nucleus in human primary hepatocytes. dl5016 0-6 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 31526233-6 2019 On the other hand, treatment of HepaRG cells with RMP resulted in a significant increase in mRNA expression for CYP2B6 (6.68-fold, p = 0.007) and CYP3A4 (111.96-fold, p = 0.001), whereas NR1I3 expression decreased (0.46-fold, p = 0.033). Rifampin 50-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-118 31218720-0 2019 The importance of both CYP2C19 and CYP2B6 germline variations in cyclophosphamide pharmacokinetics and clinical outcomes. Cyclophosphamide 65-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 31299074-6 2019 RESULTS: Within the control group, CYP2B6 983 T>C was associated with 27% higher etonogestrel Cmax and 28% higher AUC0-24weeks. etonogestrel 81-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 31299074-7 2019 In the efavirenz group CYP2B6 516 G>T was associated with 43% lower etonogestrel Cmin and 34% lower AUC0-24weeks. efavirenz 7-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 31299074-7 2019 In the efavirenz group CYP2B6 516 G>T was associated with 43% lower etonogestrel Cmin and 34% lower AUC0-24weeks. etonogestrel 68-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 31299074-10 2019 Efavirenz plus the etonogestrel contraceptive implant results in a detrimental drug-drug interaction irrespective of patient genetics, which is worsened in women possessing variant alleles for these CYP2B6 SNPs. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-205 31299074-10 2019 Efavirenz plus the etonogestrel contraceptive implant results in a detrimental drug-drug interaction irrespective of patient genetics, which is worsened in women possessing variant alleles for these CYP2B6 SNPs. etonogestrel 19-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-205 31465819-13 2019 The treatment of hepatocytes from two male human donors with 0.5-25 muM metazachlor or 500 muM NaPB for 4 days resulted in increases in CYP2B6 and CYP3A4 mRNA levels but had no effect on hepatocyte RDS. metazachlor 72-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 30786012-6 2019 Finally, the multivariable regression analysis showed that formation of tamoxifen metabolites was independently impacted by CYP2D6 diplotype and CYP3A4*22, CYP2C19*2, and CYP2B6*6 genetic polymorphisms. Tamoxifen 72-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 31243456-2 2019 Our objectives are to characterize the effects of rifampicin- and isoniazid-containing anti-TB therapy and CYP2B6 activity on efavirenz concentrations in children, using non-linear mixed-effects modelling. efavirenz 126-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 31270214-5 2019 In addition, the knockdown of ADAR1 resulted in the enhanced induction of CYP2B6 and CYP3A4 mRNA by 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime and phenobarbital, respectively. o-(3,4-dichlorobenzyl)oxime 161-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 31270214-5 2019 In addition, the knockdown of ADAR1 resulted in the enhanced induction of CYP2B6 and CYP3A4 mRNA by 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime and phenobarbital, respectively. Phenobarbital 193-206 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 30973652-7 2019 Methadone exposure was reduced when it was coadministered with CYP2B6 inducers. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 31433338-7 2019 The isoforms CYP2D6, CYP2C19, CYP2B6, and CYP2J2 are inhibited by THC and CBD. Dronabinol 66-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 31433338-7 2019 The isoforms CYP2D6, CYP2C19, CYP2B6, and CYP2J2 are inhibited by THC and CBD. Cannabidiol 74-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 31181217-4 2019 Phenanthrene, but not anthracene, significantly induced promoter activity and gene expression of human drug metabolizing enzyme CYP2B6 in HepG2 cells and human primary hepatocytes, respectively. phenanthrene 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-134 31436536-3 2019 Here, we show that in a human CAR (hCAR)-knockout (KO) HepaRG cell line, PB significantly induces the expression of CYP2B6 and CYP3A4, two shared target genes of hCAR and human PXR (hPXR). Phenobarbital 73-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 31241542-1 2019 BACKGROUND: CYP2B6 516 genotype-directed dosing improves efavirenz (EFV) exposures in HIV-infected children younger than 36 months, but such data are lacking in those with tuberculosis (TB) coinfection. efavirenz 57-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 31241542-1 2019 BACKGROUND: CYP2B6 516 genotype-directed dosing improves efavirenz (EFV) exposures in HIV-infected children younger than 36 months, but such data are lacking in those with tuberculosis (TB) coinfection. efavirenz 68-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 30973652-9 2019 In conclusion, CYP2B6 plays a prominent role in methadone metabolism, although methadone exposure is not sensitive to CYP3A perturbation. Methadone 48-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 30973652-10 2019 In designing methadone DDI studies, (1) measuring R- and S-methadone is more informative than measuring total methadone, and (2) CYP2B6 genotyping of subjects enrolled in methadone DDI studies should be considered. Methadone 13-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 30907440-0 2019 Relevance of CYP2B6 and CYP2D6 genotypes to methadone pharmacokinetics and response in the OPAL study. Methadone 44-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 31147443-1 2019 Cytochrome P450 (P450) enzymes are major catalysts involved in the oxidations of most drugs, steroids, carcinogens, fat-soluble vitamins, and natural products. Steroids 93-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 30907440-1 2019 AIMS: Our study aimed to evaluate the impacts of the cytochrome P450 (CYP) 2B6-G516T and CYP2D6 genetic polymorphisms on pharmacokinetic and clinical parameters in patients receiving methadone maintenance treatment. Methadone 183-192 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-78 30907440-4 2019 RESULTS: When comparing the three CYP2B6 genotype groups, the methadone (R)- and (S)-methadone enantiomer concentrations/doses (concentrations relative to doses) were different (P = .029, P = .0019). Methadone 62-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 30907440-4 2019 RESULTS: When comparing the three CYP2B6 genotype groups, the methadone (R)- and (S)-methadone enantiomer concentrations/doses (concentrations relative to doses) were different (P = .029, P = .0019). Arginine 73-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 30907440-4 2019 RESULTS: When comparing the three CYP2B6 genotype groups, the methadone (R)- and (S)-methadone enantiomer concentrations/doses (concentrations relative to doses) were different (P = .029, P = .0019). Methadone 81-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 30907440-7 2019 CONCLUSION: The genotyping of CYP2B6 G516T could be an interesting tool to explore methadone intervariability. Methadone 83-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 31068367-6 2019 Advantageously for its use in pharmacotherapy, alectinib further exhibited negligible potential to cause any changes in expression of ABCB1, ABCG2, ABCC1, CYP1A2, CYP3A4, and CYP2B6 in intestine, liver, and NSCLC models. alectinib 47-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 175-181 31068367-5 2019 To describe the pharmacokinetic interaction profile of alectinib in a complete fashion, its possible inhibitory properties toward clinically relevant P450 enzymes (i.e., CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, or CYP3A5) were evaluated using human P450-expressing insect microsomes, revealing alectinib as a poor interactor. alectinib 55-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-184 30048196-1 2019 The roles of human cytochrome P450 (P450 or CYP) 2A6 in the oxidation of flavanone [(2R)- and (2S)-enantiomers] and flavone were studied in human liver microsomes and recombinant human P450 enzymes. flavanone 73-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-52 30564916-10 2019 Significant contribution to the in vitro metabolism of THJ-018 and THJ-2201 originated from CYP2B6, CYP2C19, CYP3A4, and CYP3A5. thj 55-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 30564916-10 2019 Significant contribution to the in vitro metabolism of THJ-018 and THJ-2201 originated from CYP2B6, CYP2C19, CYP3A4, and CYP3A5. thj 67-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 31072872-1 2019 Cytochrome P450 (P450, CYP) enzymes are the major catalysts involved in the oxidation of steroids as well as many other compounds. Steroids 89-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-26 30048196-7 2019 CYP1A2 and CYP2B6 were also found to play significant roles in some of the oxidations of these flavonoids by human liver microsomes. Flavonoids 95-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 31213765-3 2019 Tramadol is metabolized to M1 mainly by the cytochrome P450 (CYP) 2D6 enzyme, and to other metabolites by CYP3A4 and CYP2B6. Tramadol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 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 2 subfamily B member 6 Homo sapiens 197-208 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 2 subfamily B member 6 Homo sapiens 197-208 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 2 subfamily B member 6 Homo sapiens 197-208 31226809-6 2019 Furthermore, the hepatogenic medium + NaBu pre-treatment up-regulated hepatoblast (AFP and HNF3beta) and hepatic (CK18 and ALB) markers, and increased the proportion of mature hepatocyte functions, including G6P, C/EBPalpha, and CYP2B6 mRNAs, glycogen storage and urea secretion. sethoxydim 38-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 229-235 31312405-5 2019 DL5050 preferentially induced the expression of CYP2B6 (target of hCAR) over CYP3A4 (target of hPXR) on both the mRNA and protein levels. CHEMBL4465050 0-6 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-54 30520061-6 2019 The relative induction ratios of DHI on CYP1A2, CYP2B6 and CYP3A4 activity were calculated by LC-MS/MS. dehydrosoyasaponin I 33-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-54 31022888-3 2019 Coumarins, phytochemicals abundant in food and commonly used in fragrances and cosmetics, have been shown to interact with P450 enzymes as substrates and/or inhibitors. Coumarins 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-127 30044681-3 2019 Mavacamten does not inhibit CYP enzymes, but at high concentrations relative to anticipated therapeutic concentrations induces CYP2B6 and CYP3A4 enzymes in vitro. MYK-461 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 30592088-10 2019 Exposure to phenobarbital resulted in an approximately twofold increase in CYP 2B6 enzyme activity. Phenobarbital 12-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-82 30520061-8 2019 The LC-MS/MS data showed DHI intensively inhibit CYP2A6 activity and the intensity of inhibition was followed by CYP2C8, CYP3A4, CYP2C19, CYP2B6, CYP2D6, CYP1A2, CYP2E1 and CYP2C9 in vitro. dehydrosoyasaponin I 25-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 30864294-0 2019 Occurrence of CYP2B6 516G>T polymorphism in patients with ARV-associated hepatotoxicity. omega-N-Allylarginine 61-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 30361928-6 2019 RESULTS: It was found that vonoprazan is not a significant direct inhibitor of any CYP isoforms (IC50 >= 16 muM), but shows the potential for TDI of CYP2B6, CYP2C19, and CYP3A4/5. 1-(5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine 27-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-158 30578565-0 2019 Role of CYP2B6 pharmacogenomics in bupropion-mediated smoking cessation. Bupropion 35-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 30578565-2 2019 The cytochrome P450 2B6 gene (CYP2B6) encodes CYP2B6 enzyme that has been found to mediate the hydroxylation of bupropion, a smoking cessation aid. Bupropion 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-23 30578565-2 2019 The cytochrome P450 2B6 gene (CYP2B6) encodes CYP2B6 enzyme that has been found to mediate the hydroxylation of bupropion, a smoking cessation aid. Bupropion 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 30578565-2 2019 The cytochrome P450 2B6 gene (CYP2B6) encodes CYP2B6 enzyme that has been found to mediate the hydroxylation of bupropion, a smoking cessation aid. Bupropion 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 30864294-6 2019 Likewise, hepatotoxicity in patients consuming alcohol showed higher distributions of CYP2B6 516GT, 516TT, 516GT+TT genotypes (57% vs. 25.93%; 42.86% vs. 33.33%; 71.43% vs. 59.26%). Alcohols 47-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 30864294-7 2019 Nevirapine users with hepatotoxicity overrepresented genotypes CYP2B6 TT and 516GT+TT as compared to efavirenz users (47.83% vs. 45.45%, OR = 6.88, 65.22% vs. 54.55%, OR = 1.56). Nevirapine 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 30864294-8 2019 Similarly, in nevirapine +alcohol users with hepatotoxicity, the frequency of CYP2B6 516GT, 516GT+TT genotypes was higher than with nevirapine +alcohol nonusers (40.0% vs. 11.11%, OR = 8.00, 80.0% vs. 27.78%, OR = 4.00). Nevirapine 14-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 30864294-8 2019 Similarly, in nevirapine +alcohol users with hepatotoxicity, the frequency of CYP2B6 516GT, 516GT+TT genotypes was higher than with nevirapine +alcohol nonusers (40.0% vs. 11.11%, OR = 8.00, 80.0% vs. 27.78%, OR = 4.00). Alcohols 26-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 30864294-10 2019 Likewise, in HIV patients, genotypes CYP2B6 516GT, 516GT+TT were predominant with nevirapine +alcohol users as compared to nevirapine +alcohol nonusers (57.89% vs. 34.57%, OR = 2.46; 78.95% vs. 69.14%, OR = 1.67). Nevirapine 82-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 30864294-10 2019 Likewise, in HIV patients, genotypes CYP2B6 516GT, 516GT+TT were predominant with nevirapine +alcohol users as compared to nevirapine +alcohol nonusers (57.89% vs. 34.57%, OR = 2.46; 78.95% vs. 69.14%, OR = 1.67). Alcohols 94-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 30864294-10 2019 Likewise, in HIV patients, genotypes CYP2B6 516GT, 516GT+TT were predominant with nevirapine +alcohol users as compared to nevirapine +alcohol nonusers (57.89% vs. 34.57%, OR = 2.46; 78.95% vs. 69.14%, OR = 1.67). Nevirapine 123-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 30919109-10 2019 These data indicate that PXR plays a central role in cortisol-mediated induction of CYP3A activity during pregnancy and suggests that other enzymes and transporters, such as CYP2B6 and P-glycoprotein, may also be induced during pregnancy via the same mechanism(s). Hydrocortisone 53-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 174-180 30578565-3 2019 CYP2B6 exhibits a range of polymorphic variants that alter the pharmacokinetics and pharmacodynamics of bupropion. Bupropion 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30578565-4 2019 Genetic variations in CYP2B6 may influence the risk of adverse effects or efficacy of treatment with bupropion. Bupropion 101-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 30578565-9 2019 RESULTS: There is strong literature evidence to prove that CYP2B6 polymorphisms affect pharmacokinetic and pharmacodynamic properties of bupropion, thus affecting the therapeutic outcome of smoking cessation therapy. Bupropion 137-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 31001351-6 2019 CYP3A4 was more strongly inhibited by the extract in the presence of NADPH, suggesting that the extract may inhibit CYP2B6 and CYP3A4 via mechanism-based inactivation. NADP 69-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 31001351-8 2019 Kushenol I, leachianone A, and sophoraflavone G inhibited CYP2B6, whereas kushenol C, kushenol I, kushenol M, leachianone A, and sophoraflavone G inhibited CYP3A4 via mechanism-based inhibition. kushenol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 31001351-8 2019 Kushenol I, leachianone A, and sophoraflavone G inhibited CYP2B6, whereas kushenol C, kushenol I, kushenol M, leachianone A, and sophoraflavone G inhibited CYP3A4 via mechanism-based inhibition. sophoraflavone 31-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 30906561-0 2019 Nicotine oxidation by genetic variants of CYP2B6 and in human brain microsomes. Nicotine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 30906561-2 2019 CYP2B6 is a minor contributor to hepatic nicotine metabolism, but the enzyme has been proposed as relevant to nicotine-related behaviors because of reported CYP2B6 mRNA expression in human brain tissue. Nicotine 41-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30906561-2 2019 CYP2B6 is a minor contributor to hepatic nicotine metabolism, but the enzyme has been proposed as relevant to nicotine-related behaviors because of reported CYP2B6 mRNA expression in human brain tissue. Nicotine 110-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 30906561-3 2019 Therefore, we hypothesized that CYP2B6 variants would be associated with altered nicotine oxidation, and that nicotine metabolism by CYP2B6 would be detected in human brain microsomes. Nicotine 81-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 30906561-3 2019 Therefore, we hypothesized that CYP2B6 variants would be associated with altered nicotine oxidation, and that nicotine metabolism by CYP2B6 would be detected in human brain microsomes. Nicotine 110-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 30906561-4 2019 We generated recombinant enzymes in insect cells corresponding to nine common CYP2B6 haplotypes and demonstrate genetically determined differences in nicotine oxidation to nicotine iminium ion and nornicotine for both (S) and (R)-nicotine. Nicotine 150-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 30906561-4 2019 We generated recombinant enzymes in insect cells corresponding to nine common CYP2B6 haplotypes and demonstrate genetically determined differences in nicotine oxidation to nicotine iminium ion and nornicotine for both (S) and (R)-nicotine. Nicotine 172-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 30906561-4 2019 We generated recombinant enzymes in insect cells corresponding to nine common CYP2B6 haplotypes and demonstrate genetically determined differences in nicotine oxidation to nicotine iminium ion and nornicotine for both (S) and (R)-nicotine. nornicotine 197-208 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 30906561-4 2019 We generated recombinant enzymes in insect cells corresponding to nine common CYP2B6 haplotypes and demonstrate genetically determined differences in nicotine oxidation to nicotine iminium ion and nornicotine for both (S) and (R)-nicotine. Nicotine 172-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 30906561-8 2019 Furthermore, metabolism of common CYP2B6 probe substrates, methadone and ketamine, is not detected in the presence of brain microsomes. Methadone 59-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 30906561-8 2019 Furthermore, metabolism of common CYP2B6 probe substrates, methadone and ketamine, is not detected in the presence of brain microsomes. Ketamine 73-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 30906561-9 2019 We conclude that CYP2B6 metabolizes nicotine stereoselectively and common CYP2B6 variants differ in nicotine metabolism activity, but did not find evidence of CYP2B6 activity in human brain. Nicotine 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 30906561-9 2019 We conclude that CYP2B6 metabolizes nicotine stereoselectively and common CYP2B6 variants differ in nicotine metabolism activity, but did not find evidence of CYP2B6 activity in human brain. Nicotine 100-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 30906561-9 2019 We conclude that CYP2B6 metabolizes nicotine stereoselectively and common CYP2B6 variants differ in nicotine metabolism activity, but did not find evidence of CYP2B6 activity in human brain. Nicotine 100-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 30906561-9 2019 We conclude that CYP2B6 metabolizes nicotine stereoselectively and common CYP2B6 variants differ in nicotine metabolism activity, but did not find evidence of CYP2B6 activity in human brain. Nicotine 100-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 30659730-1 2019 The high-valence iron species (Fe(IV)=O) in the cytochrome P450 enzyme superfamily is generated via the activation of O2 , and serves as the active center of selective hydrocarbon oxidation reactions. Iron 17-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-63 30659730-1 2019 The high-valence iron species (Fe(IV)=O) in the cytochrome P450 enzyme superfamily is generated via the activation of O2 , and serves as the active center of selective hydrocarbon oxidation reactions. Oxygen 118-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-63 30659730-1 2019 The high-valence iron species (Fe(IV)=O) in the cytochrome P450 enzyme superfamily is generated via the activation of O2 , and serves as the active center of selective hydrocarbon oxidation reactions. Hydrocarbons 168-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-63 30659730-2 2019 Furthermore, P450 can employ an alternate route to produce Fe(IV)=O, even from H2 O2 without O2 activation. Hydrogen Peroxide 79-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 30659730-2 2019 Furthermore, P450 can employ an alternate route to produce Fe(IV)=O, even from H2 O2 without O2 activation. Oxygen 82-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 30535366-9 2019 CYP2B6 rs2279345 polymorphism was associated with lower plasma 7-hydroxy-efavirenz:efavirenz ratio in multivariate analyses (P < 0.05). 7-hydroxy-efavirenz 63-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30638282-2 2019 CYP enzyme inhibition assays of five predominate human P450 isozymes indicate that all compounds, except for 4-hydrazide pyridine 1c, seem to be less toxic than 4-aminopyridine. 4-hydrazide pyridine 1c 109-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 30638282-2 2019 CYP enzyme inhibition assays of five predominate human P450 isozymes indicate that all compounds, except for 4-hydrazide pyridine 1c, seem to be less toxic than 4-aminopyridine. 4-Aminopyridine 161-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 30484946-7 2019 This approach suggests the potential use of noncognate P450s to produce novel oxygenated terpenoids, or to generate a novel biosynthetic route for known terpenoids by a combinatorial biosynthesis strategy. Terpenes 89-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 30484946-7 2019 This approach suggests the potential use of noncognate P450s to produce novel oxygenated terpenoids, or to generate a novel biosynthetic route for known terpenoids by a combinatorial biosynthesis strategy. Terpenes 153-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 30535366-9 2019 CYP2B6 rs2279345 polymorphism was associated with lower plasma 7-hydroxy-efavirenz:efavirenz ratio in multivariate analyses (P < 0.05). efavirenz 73-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30823561-5 2019 The metabolism of 25B-NBF was catalyzed by CYP1A1, CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2J2, CYP3A4, and UGT2B7 enzymes. SSW3KY7SWW 18-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 30576102-0 2019 Human CYP2A6, CYP2B6, AND CYP2E1 Atropselectively Metabolize Polychlorinated Biphenyls to Hydroxylated Metabolites. Polychlorinated Biphenyls 61-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 30576102-4 2019 In silico predictions with ADMET Predictor and MetaDrug software suggested a role of CYP1A2, CYP2A6, CYP2B6, CYP2E1, and CYP3A4 in the metabolism of chiral PCBs. Polychlorinated Biphenyls 156-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 30576102-10 2019 These findings suggest that CYP2A6 and CYP2B6 play an important role in the oxidation of neurotoxic PCBs to chiral OH-PCBs in humans. Polychlorinated Biphenyls 100-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 30576102-10 2019 These findings suggest that CYP2A6 and CYP2B6 play an important role in the oxidation of neurotoxic PCBs to chiral OH-PCBs in humans. oh-pcbs 115-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 30589555-0 2019 Halogen Substitution Influences Ketamine Metabolism by Cytochrome P450 2B6: In Vitro and Computational Approaches. Halogens 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-74 30589555-0 2019 Halogen Substitution Influences Ketamine Metabolism by Cytochrome P450 2B6: In Vitro and Computational Approaches. Ketamine 32-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-74 30589555-3 2019 CYP2B6 is the major catalyst of hepatic ketamine N-demethylation and metabolism at clinically relevant concentrations. ketamine n 40-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30589555-4 2019 Numerous CYP2B6 substrates contain halogens. Halogens 35-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 30589555-5 2019 CYP2B6 readily forms halogen-protein (particularly Cl-pi) bonds, which influence substrate selectivity and active site orientation. cl-pi 51-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30589555-7 2019 This investigation evaluated halogen substitution effects on CYP2B6-catalyzed ketamine analogs N-demethylation in vitro and modeled interactions with CYP2B6 using various computational approaches. Halogens 29-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 30589555-7 2019 This investigation evaluated halogen substitution effects on CYP2B6-catalyzed ketamine analogs N-demethylation in vitro and modeled interactions with CYP2B6 using various computational approaches. Ketamine 78-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 30589555-7 2019 This investigation evaluated halogen substitution effects on CYP2B6-catalyzed ketamine analogs N-demethylation in vitro and modeled interactions with CYP2B6 using various computational approaches. Nitrogen 95-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 30589555-16 2019 The binding pocket of CYP2B6 also suggested a hydrophobic component to substrate docking, on the basis of a strong linear correlation ( R2 = 0.92) between lipophilicity ( Alog P) and metabolism (log Km) of ketamine and analogs. Ketamine 206-214 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 30472588-2 2019 In this study, we used a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and N-acetyltransferase-2 (NAT2) in plasma. Dapsone 61-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 30472588-3 2019 CYP2B6 is the main catalyst of anti-HIV efavirenz, while NAT2 is involved in antitubercular drug isoniazid metabolism. Isoniazid 97-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30472588-14 2019 CONCLUSIONS: This study advocates the use of a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and NAT2, which is important in determining therapeutic dose levels of anti-HIV and anti-TB drugs in HIV/AIDS-TB coinfections. Bupropion 59-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 30472588-14 2019 CONCLUSIONS: This study advocates the use of a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and NAT2, which is important in determining therapeutic dose levels of anti-HIV and anti-TB drugs in HIV/AIDS-TB coinfections. Losartan 70-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 30472588-14 2019 CONCLUSIONS: This study advocates the use of a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and NAT2, which is important in determining therapeutic dose levels of anti-HIV and anti-TB drugs in HIV/AIDS-TB coinfections. Dapsone 83-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 30472588-2 2019 In this study, we used a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and N-acetyltransferase-2 (NAT2) in plasma. Bupropion 37-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 29310511-2 2019 We previously reported that flavone and flavanone interact spectrally with cytochrome P450 (P450 or CYP) 2A6 and 2A13 and other human P450s and inhibit catalytic activities of these P450 enzymes. flavone 28-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-117 30678826-9 2019 Interestingly, several metabolites including the 4-hydroxyl form of CPA (4-OH-CPA) and phosphamide mustard were detected in the CYP2B6, CYP2C19, and CYP3A4 expression systems, but not in the CYP2C9 and CYP2D6 system. 4-hydroxyl 49-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-134 30678826-9 2019 Interestingly, several metabolites including the 4-hydroxyl form of CPA (4-OH-CPA) and phosphamide mustard were detected in the CYP2B6, CYP2C19, and CYP3A4 expression systems, but not in the CYP2C9 and CYP2D6 system. Dimethoate 87-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-134 30678826-13 2019 Additionally, CPA metabolites like 4-OH-CPA and phosphamide mustard produced by human CYP2B6, CYP2C9, CYP2C19, and CYP3A4 are suggested to be major determinants of micronucleus induction by CPA. Dimethoate 48-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 29448869-10 2019 Validated cocktail #1 including bupropion, chlorzoxazone, ebastine and midazolam was used to characterize CYP2B6-, 2E1-, 2J2- and 3A5-mediated metabolism in human hearts. Midazolam 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 30284758-1 2019 Inspired by enzymes such as cytochrome P-450, the study of the reactivity of metalloporphyrins continues to attract major interest in the field of homogeneous catalysis. Metalloporphyrins 77-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-44 29756345-7 2019 Hydroxylation of both bupropion enantiomers, assessed by plasma hydroxybupropion/bupropion AUC ratios and urine hydroxybupropion formation clearances, was lower in CYP2B6*6/*6 but not CYP2B6*1/*6 compared with CYP2B6*1/*1 genotypes, and numerically greater in CYP2B6*4 carriers. hydroxybupropion 112-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-170 29756345-0 2019 Common Polymorphisms of CYP2B6 Influence Stereoselective Bupropion Disposition. Bupropion 57-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 29756345-9 2019 The results show that clinical hydroxylation of both bupropion enantiomers was equivalently influenced by CYP2B6 allelic variation. Bupropion 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 29756345-2 2019 This investigation determined the influence of CYP2B6 allelic variants on clinical concentrations and metabolism of bupropion enantiomers. Bupropion 116-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 29756345-10 2019 CYP2B6 polymorphisms affect S-bupropion bioactivation, which may affect therapeutic outcomes. Bupropion 28-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 29756345-7 2019 Hydroxylation of both bupropion enantiomers, assessed by plasma hydroxybupropion/bupropion AUC ratios and urine hydroxybupropion formation clearances, was lower in CYP2B6*6/*6 but not CYP2B6*1/*6 compared with CYP2B6*1/*1 genotypes, and numerically greater in CYP2B6*4 carriers. Bupropion 22-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-170 29756345-7 2019 Hydroxylation of both bupropion enantiomers, assessed by plasma hydroxybupropion/bupropion AUC ratios and urine hydroxybupropion formation clearances, was lower in CYP2B6*6/*6 but not CYP2B6*1/*6 compared with CYP2B6*1/*1 genotypes, and numerically greater in CYP2B6*4 carriers. Bupropion 22-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-190 30285275-8 2019 It is metabolized in the liver by cytochrome P450 (CYP) isoenzymes CYP3A, CYP2C19, and CYP2B6, and its main active metabolite is N-desmethylclobazam. N-desmethylclobazam 129-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 29756345-7 2019 Hydroxylation of both bupropion enantiomers, assessed by plasma hydroxybupropion/bupropion AUC ratios and urine hydroxybupropion formation clearances, was lower in CYP2B6*6/*6 but not CYP2B6*1/*6 compared with CYP2B6*1/*1 genotypes, and numerically greater in CYP2B6*4 carriers. Bupropion 22-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-190 29756345-7 2019 Hydroxylation of both bupropion enantiomers, assessed by plasma hydroxybupropion/bupropion AUC ratios and urine hydroxybupropion formation clearances, was lower in CYP2B6*6/*6 but not CYP2B6*1/*6 compared with CYP2B6*1/*1 genotypes, and numerically greater in CYP2B6*4 carriers. Bupropion 22-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-190 29756345-7 2019 Hydroxylation of both bupropion enantiomers, assessed by plasma hydroxybupropion/bupropion AUC ratios and urine hydroxybupropion formation clearances, was lower in CYP2B6*6/*6 but not CYP2B6*1/*6 compared with CYP2B6*1/*1 genotypes, and numerically greater in CYP2B6*4 carriers. hydroxybupropion 64-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-170 29756345-7 2019 Hydroxylation of both bupropion enantiomers, assessed by plasma hydroxybupropion/bupropion AUC ratios and urine hydroxybupropion formation clearances, was lower in CYP2B6*6/*6 but not CYP2B6*1/*6 compared with CYP2B6*1/*1 genotypes, and numerically greater in CYP2B6*4 carriers. Bupropion 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-170 30488807-11 2019 Activation of CYP2B6 with adaptaquin and its variants points to a potential increase in Tylenol toxicity if administered together. Adaptaquin 26-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 30488807-11 2019 Activation of CYP2B6 with adaptaquin and its variants points to a potential increase in Tylenol toxicity if administered together. Acetaminophen 88-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 30239753-0 2019 High efavirenz serum concentrations in TB/HIV-coinfected Ugandan adults with a CYP2B6 516 TT genotype on anti-TB treatment. Terbium 39-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 30320358-9 2019 Our results indicate that by stimulating HIF-1alpha activity, hypoxia downregulates the expression of CYP2B6, CYP3A4 and CYP3A5, that in turn leads to decreased conversion of CPA and IFA into their active forms and thus to diminished cytotoxicity. Cyclophosphamide 175-178 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 31339834-1 2019 PURPOSE: Hydroxylation activity at the 6beta-position of steroid hormones (testosterone, progesterone, and cortisol) by human cytochromes P450 (P450 or CYP) 3A4 and CYP3A5 and their molecular docking energy values were compared to understand the catalytic properties of the major forms of human CYP3A, namely, CYP3A4 and CYP3A5. Steroids 57-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-160 31339834-1 2019 PURPOSE: Hydroxylation activity at the 6beta-position of steroid hormones (testosterone, progesterone, and cortisol) by human cytochromes P450 (P450 or CYP) 3A4 and CYP3A5 and their molecular docking energy values were compared to understand the catalytic properties of the major forms of human CYP3A, namely, CYP3A4 and CYP3A5. Testosterone 75-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-160 31339834-1 2019 PURPOSE: Hydroxylation activity at the 6beta-position of steroid hormones (testosterone, progesterone, and cortisol) by human cytochromes P450 (P450 or CYP) 3A4 and CYP3A5 and their molecular docking energy values were compared to understand the catalytic properties of the major forms of human CYP3A, namely, CYP3A4 and CYP3A5. Progesterone 89-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-160 31339834-1 2019 PURPOSE: Hydroxylation activity at the 6beta-position of steroid hormones (testosterone, progesterone, and cortisol) by human cytochromes P450 (P450 or CYP) 3A4 and CYP3A5 and their molecular docking energy values were compared to understand the catalytic properties of the major forms of human CYP3A, namely, CYP3A4 and CYP3A5. Hydrocortisone 107-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-160 30320358-9 2019 Our results indicate that by stimulating HIF-1alpha activity, hypoxia downregulates the expression of CYP2B6, CYP3A4 and CYP3A5, that in turn leads to decreased conversion of CPA and IFA into their active forms and thus to diminished cytotoxicity. Ifosfamide 183-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 30394306-6 2019 We found that only polymethoxylated stilbenes are prone to significantly induce CYP2B6 or CYP3A4 in primary human hepatocytes via pregnane X receptor (PXR) interaction. Stilbenes 36-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 30138636-5 2018 Apatinib was a weak inhibitor of human CYP2E1 (IC50>10 muM) but inhibited CYP2B6/2B1 and CYP2D6/2D1 in a competitive way (Ki = 3.84/0.59 and 5.41/0.87 muM), and inhibited CYP3A4/3A2 and rat CYP2E1 in a mixed way (Ki = 11.50/1.83 and 13.06 muM). apatinib 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-87 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 2 subfamily B member 6 Homo sapiens 96-102 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 2 subfamily B member 6 Homo sapiens 96-102 30205091-3 2018 In vivo, polymorphism effects on methadone systemic exposure have been noted for CYP2B6, CYP3A4, and CYP2D6. Methadone 33-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 30380747-6 2018 OFIE, hydrolyzed (hdl) OFIE, and several flavonols induced the transcriptional activities of both CYP2B6 and CYP3A4 genes in HepG2 cells. Flavonols 41-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 30364229-3 2018 We found that leflunomide (LEF) and its main metabolite teriflunomide (TER), both used for autoimmune diseases treatment, induce the prototype CAR target gene CYP2B6 in primary human hepatocytes. Leflunomide 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 30364229-3 2018 We found that leflunomide (LEF) and its main metabolite teriflunomide (TER), both used for autoimmune diseases treatment, induce the prototype CAR target gene CYP2B6 in primary human hepatocytes. Leflunomide 27-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 30364229-3 2018 We found that leflunomide (LEF) and its main metabolite teriflunomide (TER), both used for autoimmune diseases treatment, induce the prototype CAR target gene CYP2B6 in primary human hepatocytes. teriflunomide 56-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 30364229-3 2018 We found that leflunomide (LEF) and its main metabolite teriflunomide (TER), both used for autoimmune diseases treatment, induce the prototype CAR target gene CYP2B6 in primary human hepatocytes. teriflunomide 71-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 30085944-0 2018 Stereoselective Ketamine Metabolism by Genetic Variants of Cytochrome P450 CYP2B6 and Cytochrome P450 Oxidoreductase. Ketamine 16-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 30085944-16 2018 CYP2B6 variants had more influence than POR variants on ketamine metabolism. Ketamine 56-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30085944-1 2018 WHAT WE ALREADY KNOW ABOUT THIS TOPIC: WHAT THIS ARTICLE TELLS US THAT IS NEW: BACKGROUND:: Human ketamine N-demethylation to norketamine in vitro at therapeutic concentrations is catalyzed predominantly by the cytochrome P4502B6 isoform (CYP2B6). Ketamine 98-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 239-245 30085944-18 2018 CONCLUSIONS: Genetic variants of CYP2B6 and P450 oxidoreductase have diminished ketamine N-demethylation activity, without affecting the stereoselectivity of metabolism. Ketamine 80-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 30085944-3 2018 CYP2B6.6, the protein encoded by the common variant allele CYP2B6*6, exhibits diminished ketamine metabolism in vitro compared with wild-type CYP2B6.1. Ketamine 89-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30085944-19 2018 These results suggest candidate genetic polymorphisms of CYP2B6 and P450 oxidoreductase for clinical evaluation to assess consequences for ketamine pharmacokinetics, elimination, bioactivation, and therapeutic effects. Ketamine 139-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 30085944-3 2018 CYP2B6.6, the protein encoded by the common variant allele CYP2B6*6, exhibits diminished ketamine metabolism in vitro compared with wild-type CYP2B6.1. Ketamine 89-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 30085944-3 2018 CYP2B6.6, the protein encoded by the common variant allele CYP2B6*6, exhibits diminished ketamine metabolism in vitro compared with wild-type CYP2B6.1. Ketamine 89-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 30085944-5 2018 This investigation evaluated ketamine metabolism by genetic variants of human CYP2B6 and POR. Ketamine 29-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). Cyclophosphamide 155-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-45 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). Cyclophosphamide 155-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). Bupropion 173-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-45 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). Bupropion 173-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). Methadone 184-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-45 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). Methadone 184-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). efavirenz 198-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-45 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). efavirenz 198-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). efavirenz 209-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-45 30201214-1 2018 Genetic variations within cytochrome P450 2B6 (CYP2B6) contribute to inter-individual variation in the metabolism of clinically important drugs, including cyclophosphamide, bupropion, methadone and efavirenz (EFZ). efavirenz 209-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 30201214-4 2018 The concentrations of CYP2B6 variant holo-enzymes were measured by using carbon monoxide (CO)-reduced difference spectroscopy, and the wild-type and 28 variants showed a peak at 450 nm. Carbon Monoxide 73-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 30201214-4 2018 The concentrations of CYP2B6 variant holo-enzymes were measured by using carbon monoxide (CO)-reduced difference spectroscopy, and the wild-type and 28 variants showed a peak at 450 nm. Carbon Monoxide 90-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 29855606-5 2018 Composite CYP2B-mediated EFV metabolism was significantly associated with CNS toxicity (p = 0.04), with extensive metabolizers reporting more and slow and very slow metabolizers reporting less toxicity after 1 month compared to intermediate metabolizers. efavirenz 25-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-15 30091858-3 2018 A mechanistic model was implemented to account for reversible and time-dependent hepatic CYP2B6 inactivation by ticlopidine, which causes elevated S-ketamine exposure in vivo. Ticlopidine 112-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-95 30091858-6 2018 Our final model produces biologically plausible output and demonstrates that ticlopidine is a strong inhibitor of CYP2B6 mediated S-ketamine metabolism. Ticlopidine 77-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 29933228-0 2018 In vitrometabolic mapping of neobavaisoflavone in human cytochromes P450 and UDP-glucuronosyltransferase enzymes by ultra high-performance liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry. neobavaisoflavone 29-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 29943426-2 2018 In humans, several P450 isoforms constitute the largest part of phase I metabolizing enzymes and catalyze oxidation reactions which convert lipophilic xenobiotics, including drugs, to more water soluble species. Water 189-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 29943426-6 2018 This P450 was identified by screening of actinobacterial strains for amodiaquine and ritonavir metabolizing activities, followed by genome sequencing and expression of the annotated S. platensis P450s in Escherichia coli. Amodiaquine 69-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 29943426-6 2018 This P450 was identified by screening of actinobacterial strains for amodiaquine and ritonavir metabolizing activities, followed by genome sequencing and expression of the annotated S. platensis P450s in Escherichia coli. Ritonavir 85-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 28876959-6 2018 Furthermore, the metabolism studies also suggested specific involvement of CYP2B6 pathway in the stereoselective hydroxylation of bupropion to R,R-hydroxybupropion, which was considered as a better marker for CYP2B6 activity. Bupropion 130-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 28876959-6 2018 Furthermore, the metabolism studies also suggested specific involvement of CYP2B6 pathway in the stereoselective hydroxylation of bupropion to R,R-hydroxybupropion, which was considered as a better marker for CYP2B6 activity. Bupropion 130-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 209-215 28876959-6 2018 Furthermore, the metabolism studies also suggested specific involvement of CYP2B6 pathway in the stereoselective hydroxylation of bupropion to R,R-hydroxybupropion, which was considered as a better marker for CYP2B6 activity. r,r-hydroxybupropion 143-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 28876959-6 2018 Furthermore, the metabolism studies also suggested specific involvement of CYP2B6 pathway in the stereoselective hydroxylation of bupropion to R,R-hydroxybupropion, which was considered as a better marker for CYP2B6 activity. r,r-hydroxybupropion 143-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 209-215 29870591-0 2018 Amenamevir: Studies of Potential CYP2C8- and CYP2B6-Mediated Pharmacokinetic Interactions With Montelukast and Bupropion in Healthy Volunteers. ASP2151 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 29870591-0 2018 Amenamevir: Studies of Potential CYP2C8- and CYP2B6-Mediated Pharmacokinetic Interactions With Montelukast and Bupropion in Healthy Volunteers. montelukast 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 29870591-0 2018 Amenamevir: Studies of Potential CYP2C8- and CYP2B6-Mediated Pharmacokinetic Interactions With Montelukast and Bupropion in Healthy Volunteers. Bupropion 111-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 29870591-1 2018 Amenamevir (formerly ASP2151) induces cytochrome P450 (CYP)2B6 and CYP3A4 and inhibits CYP2C8. ASP2151 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-62 29870591-1 2018 Amenamevir (formerly ASP2151) induces cytochrome P450 (CYP)2B6 and CYP3A4 and inhibits CYP2C8. ASP2151 21-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-62 29868865-12 2018 Conclusions: In a highly admixed Brazilian cohort, the CYP2B6 slow metabolizer genotype was associated with an increased risk of efavirenz adverse effects. efavirenz 129-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 29735754-5 2018 Conversely, PT-GP and PT-Ac caused direct inhibition of almost all P450 and UGT enzymes, with CYP1A2 (IC50 values of 0.8-4.2 muM), CYP2C8 (IC50 values of 1.1-12 muM), and UGT1A1 (IC50 values of 4.5-5.4 muM) inhibited to the greatest extent. pt-gp 12-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-79 32025958-0 2018 Effect of sex and polymorphisms of CYP2B6 and UGT1A9 on the difference between the target-controlled infusion predicted and measured plasma propofol concentration. Propofol 140-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 32025958-1 2018 INTRODUCTION: To examine whether sex and polymorphisms of cytochrome P450 (CYP) 2B6 and UDP-glucuronosyltransferase (UGT) 1A9 affect the difference between predicted and measured plasma propofol concentration during continuous infusion by target-controlled infusion. Propofol 186-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-83 29735754-5 2018 Conversely, PT-GP and PT-Ac caused direct inhibition of almost all P450 and UGT enzymes, with CYP1A2 (IC50 values of 0.8-4.2 muM), CYP2C8 (IC50 values of 1.1-12 muM), and UGT1A1 (IC50 values of 4.5-5.4 muM) inhibited to the greatest extent. 6-(3-propylthio-1,2,5-thiadiazol-4-yl)-1-azabicyclo(3.2.1)octane 22-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-79 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 2 subfamily B member 6 Homo sapiens 99-105 30159225-1 2018 Objectives: The CYP2B6 is one of the most polymorphic CYP genes in humans that has the potential to modify the pharmacological and toxicological responses to clinically important drugs such as antimalarial artemisinin and its derivatives. artemisinin 206-217 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 30159225-2 2018 The aim of the study was to determine the frequency of CYP2B6 polymorphisms in Timor malaria endemic area, East Nusa Tenggara, Indonesia where Artemisin-based Combination Therapy (ACT) has been used to treat uncomplicated malaria. artemisin 143-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 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 2 subfamily B member 6 Homo sapiens 81-87 29695615-7 2018 In human hepatocytes, radiometric integration attributed 43% of the total metabolism of SAM-760 to this non-P450 pathway. UNII-4JEH3BL51N 88-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 29695615-10 2018 We also determined the relative contribution of P450 to the metabolism of SAM-760 in human hepatocytes by following the rate of formation of oxidative metabolites in the presence and absence of P450 isoform-specific inhibitors. UNII-4JEH3BL51N 74-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 29695615-11 2018 The P450-mediated oxidative metabolism of SAM-760 was still primarily attributed to CYP3A (33%), with minor contributions from P450 isoforms CYP2C19 and CYP2D6. UNII-4JEH3BL51N 42-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-8 29458047-6 2018 Several SNPs in the CYP2B6, 3A4, 2C19, 2D6, and 3A5 genes result in increases in methadone plasma concentrations, decreased N-demethylation, and decreased methadone clearance. Methadone 81-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 29458047-6 2018 Several SNPs in the CYP2B6, 3A4, 2C19, 2D6, and 3A5 genes result in increases in methadone plasma concentrations, decreased N-demethylation, and decreased methadone clearance. Methadone 155-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 29458047-7 2018 In particular, carriers of CYP2B6*6/*6 may have a greater risk for detrimental adverse effects, as methadone metabolism and clearance are diminished in these individuals. Methadone 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 29458047-8 2018 CYP2B6*4, on the other hand, has been observed to decrease plasma concentrations of methadone due to increased methadone clearance. Methadone 84-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 29458047-8 2018 CYP2B6*4, on the other hand, has been observed to decrease plasma concentrations of methadone due to increased methadone clearance. Methadone 111-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 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 2 subfamily B member 6 Homo sapiens 69-75 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 2 subfamily B member 6 Homo sapiens 69-75 29914177-10 2018 Induction of the CYP2B6 in pediatric gliomas with lower expression of the enzyme, could be an alternative to improve the antitumoral effect of CPA treatment. Cyclophosphamide 143-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 28737446-9 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.24, 0.28, 0.15, 0.45, 0.46, 0.42 and 0.54, respectively, in HLM determined by chemical inhibition method. Clopidogrel 50-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-9 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.24, 0.28, 0.15, 0.45, 0.46, 0.42 and 0.54, respectively, in HLM determined by chemical inhibition method. Ketamine 63-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-9 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.24, 0.28, 0.15, 0.45, 0.46, 0.42 and 0.54, respectively, in HLM determined by chemical inhibition method. Selegiline 73-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-9 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.24, 0.28, 0.15, 0.45, 0.46, 0.42 and 0.54, respectively, in HLM determined by chemical inhibition method. Sertraline 85-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-9 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.24, 0.28, 0.15, 0.45, 0.46, 0.42 and 0.54, respectively, in HLM determined by chemical inhibition method. Ticlopidine 100-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-10 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.46, 0.17, 0.15, 0.60, 0.51, 0.66 and 0.77, respectively, in HLM determined by chemical inhibition method in the presence of BSA (0.5% w/v). artemisinin 26-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-10 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.46, 0.17, 0.15, 0.60, 0.51, 0.66 and 0.77, respectively, in HLM determined by chemical inhibition method in the presence of BSA (0.5% w/v). Bupropion 39-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-10 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.46, 0.17, 0.15, 0.60, 0.51, 0.66 and 0.77, respectively, in HLM determined by chemical inhibition method in the presence of BSA (0.5% w/v). Clopidogrel 50-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-10 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.46, 0.17, 0.15, 0.60, 0.51, 0.66 and 0.77, respectively, in HLM determined by chemical inhibition method in the presence of BSA (0.5% w/v). Ketamine 63-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-10 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.46, 0.17, 0.15, 0.60, 0.51, 0.66 and 0.77, respectively, in HLM determined by chemical inhibition method in the presence of BSA (0.5% w/v). Selegiline 73-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-10 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.46, 0.17, 0.15, 0.60, 0.51, 0.66 and 0.77, respectively, in HLM determined by chemical inhibition method in the presence of BSA (0.5% w/v). Sertraline 85-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-10 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.46, 0.17, 0.15, 0.60, 0.51, 0.66 and 0.77, respectively, in HLM determined by chemical inhibition method in the presence of BSA (0.5% w/v). Ticlopidine 100-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-13 2018 Ticlopidine is a time-dependent inhibitor of both CYP2B6 and CYP2C19 that can inhibit the bupropion metabolism by 50-60%. Ticlopidine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 28737446-13 2018 Ticlopidine is a time-dependent inhibitor of both CYP2B6 and CYP2C19 that can inhibit the bupropion metabolism by 50-60%. Bupropion 90-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 29635009-0 2018 The impact of CYP2B6 polymorphisms on the interactions of efavirenz with lumefantrine: Implications for paediatric antimalarial therapy. efavirenz 58-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 29635009-0 2018 The impact of CYP2B6 polymorphisms on the interactions of efavirenz with lumefantrine: Implications for paediatric antimalarial therapy. Lumefantrine 73-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 29635009-2 2018 The concomitant use of lumefantrine with the antiretroviral efavirenz, which is metabolised by CYP2B6 and is an inducer of CYP3A4, increases the risk of lumefantrine failure and can result in an increased recrudescence rate in HIV-infected children. Lumefantrine 23-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-101 29635009-3 2018 This is further confounded by CYP2B6 being highly polymorphic resulting in a 2-3 fold higher efavirenz plasma concentration in polymorphic subjects, which enhances the potential for an efavirenz-lumefantrine drug-drug interaction (DDI). efavirenz-lumefantrine 185-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 29689453-3 2018 In order to understand the impact of phytocannabinoids on human cardiovascular health, there is a need to study the metabolism of phytocannabinoids by cardiac cytochromes p450 (CYPs). phytocannabinoids 37-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-175 29689453-3 2018 In order to understand the impact of phytocannabinoids on human cardiovascular health, there is a need to study the metabolism of phytocannabinoids by cardiac cytochromes p450 (CYPs). phytocannabinoids 130-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-175 28737446-9 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.24, 0.28, 0.15, 0.45, 0.46, 0.42 and 0.54, respectively, in HLM determined by chemical inhibition method. artemisinin 26-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 28737446-9 2018 The fm, CYP2B6 values for artemisinin, bupropion, clopidogrel, ketamine, selegiline, sertraline and ticlopidine were 0.24, 0.28, 0.15, 0.45, 0.46, 0.42 and 0.54, respectively, in HLM determined by chemical inhibition method. Bupropion 39-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 29842871-10 2018 We found that the metabolism of fluorescent substrates by CYP2B6, CYP2C19, CYP2E1, and CYP1A1 in the presence of serum obtained from DSS-treated mice was activated by 42%, 37%, 37%, and 23%, respectively, relative to that associated with sera from control mice. 3-(trimethylsilyl)propanesulfonic acid 133-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 29621538-9 2018 These results suggest a positive feedback loop by which estrogens can augment the effects of ketamine and its (2R,6R)-HNK and (2S,6S)-HNK metabolites on the ERalpha-induced transcription of CYP2A6 and CYP2B6, estrogen inducible enzymes that catalyze ketamine"s biotransformation to form the two active metabolites. Ketamine 93-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 201-207 29522085-0 2018 Association of CYP2B6 Single-Nucleotide Polymorphisms Altering Efavirenz Metabolism With Hepatitis C Virus (HCV) Treatment Relapse Among Human Immunodeficiency Virus/HCV-Coinfected African Americans Receiving Ledipasvir/Sofosbuvir in the ION-4 Trial. single-nucleotide 22-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 29723517-0 2018 Inhibitory effect of alpha-terpinyl acetate on cytochrome P450 2B6 enzymatic activity. terpinyl acetate 21-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-66 29522085-0 2018 Association of CYP2B6 Single-Nucleotide Polymorphisms Altering Efavirenz Metabolism With Hepatitis C Virus (HCV) Treatment Relapse Among Human Immunodeficiency Virus/HCV-Coinfected African Americans Receiving Ledipasvir/Sofosbuvir in the ION-4 Trial. ledipasvir 209-219 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 29522085-0 2018 Association of CYP2B6 Single-Nucleotide Polymorphisms Altering Efavirenz Metabolism With Hepatitis C Virus (HCV) Treatment Relapse Among Human Immunodeficiency Virus/HCV-Coinfected African Americans Receiving Ledipasvir/Sofosbuvir in the ION-4 Trial. Sofosbuvir 220-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 29650790-11 2018 Our results indicate that oleamide caused the downregulation of some rat liver P450s, and hormones are not mediators of this effect. oleylamide 26-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-84 29650790-2 2018 We studied the influence of the endocannabinoid oleamide on rat and human liver P450s. oleylamide 48-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-85 28639016-7 2018 The docking models showed that human CYP1A1, CYP2A6, and CYP2B6 isozymes have the potential to metabolize CB118 and CB153. cb118 106-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 28639016-7 2018 The docking models showed that human CYP1A1, CYP2A6, and CYP2B6 isozymes have the potential to metabolize CB118 and CB153. 2,4,5,2',4',5'-hexachlorobiphenyl 116-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 29671087-6 2018 A number of mechanisms could be responsible for his increased sensitivity to chemicals following exposure to fluconazole/ketoconazole, including inhibition of P450 and other detoxification enzymes, acetaldehyde buildup, and neurogenic sensitization. Fluconazole 109-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-163 28648140-2 2018 1-Chloropyrene, one of the major chlorinated polycyclic aromatic hydrocarbon contaminants, was incubated with human cytochrome P450 (P450 or CYP) enzymes including CYP1A1, 1A2, 1B1, 2A6, 2A13, 2B6, 2C9, 2D6, 2E1, 3A4 and 3A5. Polycyclic Aromatic Hydrocarbons 59-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-158 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 2 subfamily B member 6 Homo sapiens 153-159 28657402-12 2018 It is recommended that olaparib is given with caution with narrow therapeutic range or sensitive CYP3A substrates, and that prescribers are aware that olaparib may reduce exposure to substrates of CYP2B6. olaparib 151-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 197-203 28730856-7 2018 Changes in mRNA expression in human hepatocytes and enzyme activity in human liver microsomes by budesonide were determined for CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and CYP3A. Budesonide 97-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 29136336-7 2018 Polymorphisms in CYP2C19 and CYP2B6 genes influenced sertraline pharmacokinetics, with a greater effect of CYP2C19. Sertraline 53-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 29136336-13 2018 There was a tendency to present more adverse drug reactions in women and individuals with higher AUC of sertraline, such as CYP2C19 intermediate metabolizers and CYP2B6 G516T T/T individuals. Sertraline 104-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 29453199-6 2018 In addition, the induction potential of RO6889678 on cytochrome P450 (P450) enzymes and transporters at steady state was assessed and cotreatment with ritonavir revealed a complex drug-drug interaction with concurrent P450 inhibition and moderate UDP-glucuronosyltransferase induction. RO6889678 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 29453199-6 2018 In addition, the induction potential of RO6889678 on cytochrome P450 (P450) enzymes and transporters at steady state was assessed and cotreatment with ritonavir revealed a complex drug-drug interaction with concurrent P450 inhibition and moderate UDP-glucuronosyltransferase induction. RO6889678 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 29453199-6 2018 In addition, the induction potential of RO6889678 on cytochrome P450 (P450) enzymes and transporters at steady state was assessed and cotreatment with ritonavir revealed a complex drug-drug interaction with concurrent P450 inhibition and moderate UDP-glucuronosyltransferase induction. RO6889678 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 29671087-6 2018 A number of mechanisms could be responsible for his increased sensitivity to chemicals following exposure to fluconazole/ketoconazole, including inhibition of P450 and other detoxification enzymes, acetaldehyde buildup, and neurogenic sensitization. Ketoconazole 121-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-163 29637923-0 2018 Effects of Genetic Polymorphisms of CYP2B6 on the Pharmacokinetics of Bupropion and Hydroxybupropion in Healthy Chinese Subjects. Bupropion 70-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 29637923-0 2018 Effects of Genetic Polymorphisms of CYP2B6 on the Pharmacokinetics of Bupropion and Hydroxybupropion in Healthy Chinese Subjects. hydroxybupropion 84-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 29454235-0 2018 Donor and recipient P450 gene polymorphisms influence individual pharmacological effects of tacrolimus in Chinese liver transplantation patients. Tacrolimus 92-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 28298143-0 2018 P450 Eicosanoids and Reactive Oxygen Species Interplay in Brain Injury and Neuroprotection. Eicosanoids 5-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 29454235-1 2018 The immunosuppressant drug tacrolimus (Tac) used for the prevention of immunological rejection is a metabolic substrate of cytochrome P450 enzymes. Tacrolimus 27-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-138 29596329-2 2018 More recently, an important role has emerged for halogen-pi interactions in the CYP2B6 active site in substrate selectivity, explaining in part the preference for halogenated ligands as substrates. Halogens 49-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 29468507-0 2018 Impact of clinical factors and UGT1A9 and CYP2B6 genotype on inter-individual differences in propofol pharmacokinetics. Propofol 93-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 29468507-6 2018 The inter-individual variability of the propofol pharmacokinetics was evaluated by investigating relationships between AUC15min and genotype of UGT1A9 and CYP2B6; clinical factors, such as age, sex, body mass index (BMI), and preoperative hematological examination; and hemodynamic variables measured by a pulse dye densitogram analyzer. Propofol 40-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-161 29596329-3 2018 The mechanism by which such ligands interact with CYP2B enzymes involves conserved phenylalanine side chains, in particular F108, F115, or F297, in the active site, which form pi bonds with halogens. Phenylalanine 83-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-55 29596329-4 2018 To illustrate such halogen-pi interactions using drugs that are major substrates of CYP2B6, we present here a crystal structure of CYP2B6 in complex with an analog of the widely used anti-HIV drug efavirenz, which contains a methyl group in place of the carbonyl oxygen. Halogens 19-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 29596329-4 2018 To illustrate such halogen-pi interactions using drugs that are major substrates of CYP2B6, we present here a crystal structure of CYP2B6 in complex with an analog of the widely used anti-HIV drug efavirenz, which contains a methyl group in place of the carbonyl oxygen. Halogens 19-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 29596329-4 2018 To illustrate such halogen-pi interactions using drugs that are major substrates of CYP2B6, we present here a crystal structure of CYP2B6 in complex with an analog of the widely used anti-HIV drug efavirenz, which contains a methyl group in place of the carbonyl oxygen. Oxygen 263-269 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 29596329-4 2018 To illustrate such halogen-pi interactions using drugs that are major substrates of CYP2B6, we present here a crystal structure of CYP2B6 in complex with an analog of the widely used anti-HIV drug efavirenz, which contains a methyl group in place of the carbonyl oxygen. Oxygen 263-269 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 29636682-1 2018 Poor metabolisers of CYP2B6 (PM) require a lower dose of efavirenz because of serious adverse reactions resulting from the higher plasma concentrations associated with a standard dose. efavirenz 57-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 29629676-2 2018 EFV is metabolised in the liver via CYP2B6, and genetic polymorphism of CYP2B6 is known to result in slowed metabolism of the drug. efavirenz 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 29629676-2 2018 EFV is metabolised in the liver via CYP2B6, and genetic polymorphism of CYP2B6 is known to result in slowed metabolism of the drug. efavirenz 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 29285606-8 2018 Tamoxifen bisphenol was mainly formed from (E)- and (Z)-metabolite E by CYP2B6 and CYP2C19, respectively. 1,1-bis(4-hydroxyphenyl)-2-phenylbut-1-ene 0-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 29371396-1 2018 Mitochondrial cytochromes P450 (P450s) are responsible for important metabolic reactions, including steps involved in steroid and vitamin D metabolism. Steroids 118-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 29371396-1 2018 Mitochondrial cytochromes P450 (P450s) are responsible for important metabolic reactions, including steps involved in steroid and vitamin D metabolism. Vitamin D 130-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 29371396-2 2018 The mitochondrial P450 24A1 (CYP24A1) is responsible for deactivation of the bioactive form of vitamin D, 1,25(OH)2D3. Vitamin D 95-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-22 29371396-5 2018 In this study, we report the results of solution NMR in which we monitor isotopically labeled full-length Adx as it binds CYP24A1 in complex with the P450 inhibitor clotrimazole. Clotrimazole 165-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 29215266-0 2018 Use of Phenoxyaniline Analogues To Generate Biochemical Insights into the Interactio n of Polybrominated Diphenyl Ether with CYP2B Enzymes. 3-(phenoxy)aminobenzene 7-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-130 30363128-7 2018 BPH with low susceptibility to etofenprox showed a marked increase in P450 activity after RRSV infection; the GST-CDNB activity of females in the insecticide-resistant strain increased. ethofenprox 31-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 30363128-8 2018 RGSV infection induced high CE and P450 activities in etofenprox-selected females. ethofenprox 54-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 29215266-0 2018 Use of Phenoxyaniline Analogues To Generate Biochemical Insights into the Interactio n of Polybrominated Diphenyl Ether with CYP2B Enzymes. Halogenated Diphenyl Ethers 90-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-130 29215266-2 2018 CYP2B6 is a major catalyst of biotransformation of environmental toxicants, including polybrominated diphenyl ethers (PBDEs). Halogenated Diphenyl Ethers 86-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 29215266-2 2018 CYP2B6 is a major catalyst of biotransformation of environmental toxicants, including polybrominated diphenyl ethers (PBDEs). Halogenated Diphenyl Ethers 118-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 29215266-3 2018 CYP2B substrates tend to contain halogen atoms, but the biochemical basis for this selectivity and for species specific determinants of metabolism has not been identified. Halogens 33-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 29215266-4 2018 Spectral binding titrations and inhibition studies were performed to investigate interactions of rat CYP2B1, rabbit CYP2B4, and CYP2B6 with a series of phenoxyaniline (POA) congeners that are analogues of PBDEs. 3-(phenoxy)aminobenzene 152-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-134 29215266-4 2018 Spectral binding titrations and inhibition studies were performed to investigate interactions of rat CYP2B1, rabbit CYP2B4, and CYP2B6 with a series of phenoxyaniline (POA) congeners that are analogues of PBDEs. phenoxyacetic acid 168-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-134 29215266-7 2018 CYP2B1 was the enzyme most sensitive to POA congeners, so the Val-363 residue from that enzyme was introduced into CYP2B4 or CYP2B6. Valine 62-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 29215266-12 2018 These results provide insight into the biochemical basis of binding of diphenyl ethers to human CYP2B6 and changes in CYP2B6-mediated metabolism that are dependent on POA congener and redox partner identity. Phenyl Ethers 71-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 29162613-4 2018 Ruxolitinib was found to fully inhibit IL-6-mediated repression of P450 (CYP1A2, CYP2B6, and CYP3A4) and transporter (NTCP, OATP1B1, and OCT1) mRNA levels in primary human hepatocytes and differentiated hepatoma HepaRG cells. ruxolitinib 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 29162613-7 2018 By contrast, ruxolitinib failed to suppress the repression of drug-detoxifying protein mRNA levels caused by IL-1beta The JAK inhibitor and anti-rheumatoid arthritis compound tofacitinib was additionally found to reverse IL-6-mediated suppression of P450 and transporter mRNA expressions. tofacitinib 175-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 250-270 29305929-4 2018 CYP2B6 played an exclusive role in the formation of 2-hydroxylauric acid. 2-Hydroxydodecanoic acid 52-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 29305929-5 2018 The production of 11-hydroxylauric acid was mediated by CYP2E1, CYP2C9, CYP2B6, CYP1A2, CYP3A4, and CYP4A11. 11-hydroxydodecanoic acid 18-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 29314710-8 2018 Second, enzyme kinetic studies showed that the initial metabolic steps were formed by cytochrome P450 isoforms (CYP) CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 resulting in pyrrolidine, thiophene or alkyl hydroxy metabolites depending on the length of the alkyl chain. pyrrolidine 182-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 29314710-8 2018 Second, enzyme kinetic studies showed that the initial metabolic steps were formed by cytochrome P450 isoforms (CYP) CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 resulting in pyrrolidine, thiophene or alkyl hydroxy metabolites depending on the length of the alkyl chain. Thiophenes 195-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 29165700-3 2018 TCDD activates cytochrome (CYP) p450 metabolic enzymes that alter organ function to cause disease. Polychlorinated Dibenzodioxins 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 29357810-6 2018 RESULTS: Although the activities of CYP2B6 and CYP3A were induced by treatment with PB and RIF, we found that the activity of phenacetin O-deethylase (PHOD), which is known as a marker of the activity of CYP1A2, was also enhanced by treatment with these non-CYP1A2 inducers in HepaRG cells. Phenobarbital 84-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 27414183-10 2017 In conclusion, TAK-438 was primarily metabolized by multiple metabolizing enzymes including CYP3A4, CYP2B6, CYP2C19, CYP2D6, and a non-CYP enzyme SULT2A1, and the influence of the CYP2C19 genotype status on gastric acid suppression post TAK-438 dosing could be small. 1-(5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine 15-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 29157826-6 2017 RESULTS: Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). Rifampin 127-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 29157826-6 2017 RESULTS: Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). Rifampin 137-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 29157826-6 2017 RESULTS: Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 165-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 29157826-10 2017 Taken together, these results show that UA inhibits the transactivation effects of PXR and CAR, and reduces the expression and function of CYP3A4 and CYP2B6. ursolic acid 40-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 27414183-6 2017 Furthermore, CYP3A4 mainly contributed to the metabolism of TAK-438 to M-I, M-III, and N-demethylated TAK-438, and CYP2B6, CYP2C19 and CYP2D6 partly catalyzed the metabolism of TAK-438. tak 60-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 29463407-0 2018 Effect of genetic polymorphism of human CYP2B6 on the metabolic activation of chlorpyrifos. Chlorpyrifos 78-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 29463407-2 2018 Chlorpyrifos oxon (CPO) is a toxic metabolite of CPS that is produced by CYP2B6. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 0-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 29463407-2 2018 Chlorpyrifos oxon (CPO) is a toxic metabolite of CPS that is produced by CYP2B6. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 19-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 29463407-2 2018 Chlorpyrifos oxon (CPO) is a toxic metabolite of CPS that is produced by CYP2B6. Chlorpyrifos 49-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 29463407-3 2018 In this study, we examined the variability of CPS metabolism resulting from single-nucleotide polymorphisms in CYP2B6. Chlorpyrifos 46-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 29463407-6 2018 The conversion of CPS to CPO by the CYP2B6 variants was analyzed with high-performance liquid chromatography. Chlorpyrifos 18-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 29463407-6 2018 The conversion of CPS to CPO by the CYP2B6 variants was analyzed with high-performance liquid chromatography. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 25-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 29463407-8 2018 The CYP2B6 variants produced CPO with the following kinetic parameters: Km for CYP2B6.5 and CYP2B6.8 were 20.44+-6.43 and 44.69+-9.97muM, respectively; and Vmax were 1.10+-0.10 and 1.77+-0.26mol/min/mol P450, respectively. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 29-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 29463407-8 2018 The CYP2B6 variants produced CPO with the following kinetic parameters: Km for CYP2B6.5 and CYP2B6.8 were 20.44+-6.43 and 44.69+-9.97muM, respectively; and Vmax were 1.10+-0.10 and 1.77+-0.26mol/min/mol P450, respectively. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 29-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 29463407-8 2018 The CYP2B6 variants produced CPO with the following kinetic parameters: Km for CYP2B6.5 and CYP2B6.8 were 20.44+-6.43 and 44.69+-9.97muM, respectively; and Vmax were 1.10+-0.10 and 1.77+-0.26mol/min/mol P450, respectively. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 29-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 29463407-9 2018 These results indicate that the amino acid substitutions in the CYP2B6 variants suppressed the metabolic activation of CPS. Chlorpyrifos 119-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 29148818-0 2017 Dissociation Constants of Cytochrome P450 2C9/Cytochrome P450 Reductase Complexes in a Lipid Bilayer Membrane Depend on NADPH: A Single-Protein Tracking Study. NADP 120-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-41 28866862-5 2017 SKF-525A, a widely used non-specific P450 inhibitor, had no inhibitory activity against CYP1A2, 2A6, 2E1 and 2J2, while it showed an inhibitory effect against CYP2B6, CYP2C19 and 2D6 with IC50 values of 2.5, 3.6 and 0.5 mu m, respectively. Proadifen 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 28699698-3 2017 This study discovered that the estrogen-response element single nucleotide polymorphism (ERE-SNP; rs16974799, C/T) of cytochrome 2B6 gene (cyp2b6; methadone catabolic enzyme) responded differently to MMT dosing. Methadone 147-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 139-145 28699698-3 2017 This study discovered that the estrogen-response element single nucleotide polymorphism (ERE-SNP; rs16974799, C/T) of cytochrome 2B6 gene (cyp2b6; methadone catabolic enzyme) responded differently to MMT dosing. mmt 200-203 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 139-145 29157826-6 2017 RESULTS: Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). ursolic acid 51-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 28941798-8 2017 In human liver microsomes, shikonin was not only a mixed inhibitor of CYP1A2, CYP2B6, CYP2C9, CYP2D6 and CYP3A4, but also a competitive inhibitor of CYP2E1, with Ki values no more than 7.72muM. shikonin 27-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 29156454-6 2017 p450 enzymes exhibit C-H oxidation site-selectivity, in which the enzyme scaffold causes a specific C-H bond to be functionalized by placing it close to the iron-oxo haem complex. Iron 157-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 29156454-6 2017 p450 enzymes exhibit C-H oxidation site-selectivity, in which the enzyme scaffold causes a specific C-H bond to be functionalized by placing it close to the iron-oxo haem complex. Heme 166-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 28947469-3 2017 We demonstrate associations between CYP2B6 polymorphisms and metabolite ratios for both 3-hydroxynevirapine and 8-hydroxynevirapine, suggesting involvement of CYP2B6 in generating these metabolites. 3-Hydroxy Nevirapine 88-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 28947469-3 2017 We demonstrate associations between CYP2B6 polymorphisms and metabolite ratios for both 3-hydroxynevirapine and 8-hydroxynevirapine, suggesting involvement of CYP2B6 in generating these metabolites. 3-Hydroxy Nevirapine 88-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 28947469-3 2017 We demonstrate associations between CYP2B6 polymorphisms and metabolite ratios for both 3-hydroxynevirapine and 8-hydroxynevirapine, suggesting involvement of CYP2B6 in generating these metabolites. 8-Hydroxynevirapine 112-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 28947469-3 2017 We demonstrate associations between CYP2B6 polymorphisms and metabolite ratios for both 3-hydroxynevirapine and 8-hydroxynevirapine, suggesting involvement of CYP2B6 in generating these metabolites. 8-Hydroxynevirapine 112-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 29133890-6 2017 Initially we found that two variants, c.516 G > T in the CYP2B6 gene and c.2677 T > G in the ABCB1 gene, significantly correlate with propofol"s metabolic profile, however after Bonferroni correction the P-values were not statistically significant. Propofol 140-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 29133890-7 2017 Our results suggest, that variants within the CYP2B6 and ABCB1 genes correlate stronger with propofol"s metabolic profile compared to other 7 genes. Propofol 93-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 28927457-3 2017 Tetracyclines and macrolides via inhibiting p450 enzymes attenuate ATRA degradation, thereby increase p53. Tetracyclines 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 28904078-2 2017 Here, we studied the interactions of human CYP2D6, a major metabolizer of psychoactive drugs, with one of the most prevalent human P450 enzymes, ethanol-inducible CYP2E1. Ethanol 145-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-135 28723731-9 2017 CONCLUSIONS: Patients homozygous for CYP2B6*6 had a >90% higher methadone C/D ratio. Methadone 67-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 28723731-10 2017 Genotyping of CYP2B6 may therefore be of value when assessing dose requirements in methadone maintenance treatment. Methadone 83-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 28887089-6 2017 In addition, the inhibitory activities of steviol and stevioside towards the major cytochrome P450 enzymes CYP3A4, CYP2C9, CYP2D6, CYP1A2 and CYP2B6 were evaluated in vitro. steviol 42-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 28887089-6 2017 In addition, the inhibitory activities of steviol and stevioside towards the major cytochrome P450 enzymes CYP3A4, CYP2C9, CYP2D6, CYP1A2 and CYP2B6 were evaluated in vitro. stevioside 54-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 28904078-9 2017 Our results further emphasize the role of P450-P450 interactions in regulatory cross-talk in human drug-metabolizing ensemble and suggest a role of interactions of CYP2E1 with CYP2D6 in pharmacologically important instances of alcohol-drug interactions. Alcohols 227-234 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 28904078-9 2017 Our results further emphasize the role of P450-P450 interactions in regulatory cross-talk in human drug-metabolizing ensemble and suggest a role of interactions of CYP2E1 with CYP2D6 in pharmacologically important instances of alcohol-drug interactions. Alcohols 227-234 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 28945086-4 2017 OA attenuated the promoter activities, expressions, and enzyme catalytic activities of CYP3A4 and CYP2B6 mediated by rifampin (RIF) and CITCO. Rifampin 117-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 28945086-4 2017 OA attenuated the promoter activities, expressions, and enzyme catalytic activities of CYP3A4 and CYP2B6 mediated by rifampin (RIF) and CITCO. Rifampin 127-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 28945086-4 2017 OA attenuated the promoter activities, expressions, and enzyme catalytic activities of CYP3A4 and CYP2B6 mediated by rifampin (RIF) and CITCO. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 136-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 28070879-8 2017 In the presence of efavirenz, mutations in CYP2B6 influenced the number of patients achieving day-7 lumefantrine concentrations above accepted therapeutic cut-offs. day-7 lumefantrine 94-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 28723731-0 2017 Combined Effect of CYP2B6 Genotype and Other Candidate Genes on a Steady-State Serum Concentration of Methadone in Opioid Maintenance Treatment. Methadone 102-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 28723731-2 2017 The aim of this study was to clarify the impact of the reduced function CYP2B6*6 variant allele together with variants in other candidate genes on a steady-state methadone concentration in a naturalistic setting. Methadone 162-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 28723731-6 2017 The estimated mean methadone C/D ratios was 17.8 nmol L mg for homozygous carriers of CYP2B6*6, which was significantly (P < 0.001) higher than noncarriers (9.2 nmol L mg). Methadone 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 28723731-6 2017 The estimated mean methadone C/D ratios was 17.8 nmol L mg for homozygous carriers of CYP2B6*6, which was significantly (P < 0.001) higher than noncarriers (9.2 nmol L mg). Magnesium 56-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 29018344-1 2017 Xanthates (alkyl or aryl derivatives of dithiocarbonic acid) have been shown to be selective mechanism-based inactivators of cytochromes P450 2B1/2B6 and 2E1 due to covalent binding of a reactive intermediate to apoprotein after double hydrogen abstraction at alpha-carbon atom, suggesting interaction of the xanthate dithiocarbonic head with the enzyme heme. thioketone 40-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-157 29018344-1 2017 Xanthates (alkyl or aryl derivatives of dithiocarbonic acid) have been shown to be selective mechanism-based inactivators of cytochromes P450 2B1/2B6 and 2E1 due to covalent binding of a reactive intermediate to apoprotein after double hydrogen abstraction at alpha-carbon atom, suggesting interaction of the xanthate dithiocarbonic head with the enzyme heme. Hydrogen 236-244 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-157 29018344-1 2017 Xanthates (alkyl or aryl derivatives of dithiocarbonic acid) have been shown to be selective mechanism-based inactivators of cytochromes P450 2B1/2B6 and 2E1 due to covalent binding of a reactive intermediate to apoprotein after double hydrogen abstraction at alpha-carbon atom, suggesting interaction of the xanthate dithiocarbonic head with the enzyme heme. Carbon 46-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-157 29018344-1 2017 Xanthates (alkyl or aryl derivatives of dithiocarbonic acid) have been shown to be selective mechanism-based inactivators of cytochromes P450 2B1/2B6 and 2E1 due to covalent binding of a reactive intermediate to apoprotein after double hydrogen abstraction at alpha-carbon atom, suggesting interaction of the xanthate dithiocarbonic head with the enzyme heme. Heme 354-358 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-157 28927457-3 2017 Tetracyclines and macrolides via inhibiting p450 enzymes attenuate ATRA degradation, thereby increase p53. Macrolides 18-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 28187506-5 2017 Efavirenz group: CYP2B6 516G>T was associated with lower levonorgestrel log10 Cmax and log10 AUC. Levonorgestrel 60-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 29302220-6 2017 In pharmacogenetics of opioid addictions, methadone dose may be regulated by variants in cytochrome P450 2B6 (CYP2B6), a methadone-metabolizing enzyme, and by a locus 300 kb 5" to OPRM1. Methadone 42-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-108 29302220-6 2017 In pharmacogenetics of opioid addictions, methadone dose may be regulated by variants in cytochrome P450 2B6 (CYP2B6), a methadone-metabolizing enzyme, and by a locus 300 kb 5" to OPRM1. Methadone 42-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 29302220-6 2017 In pharmacogenetics of opioid addictions, methadone dose may be regulated by variants in cytochrome P450 2B6 (CYP2B6), a methadone-metabolizing enzyme, and by a locus 300 kb 5" to OPRM1. Methadone 121-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-108 29302220-6 2017 In pharmacogenetics of opioid addictions, methadone dose may be regulated by variants in cytochrome P450 2B6 (CYP2B6), a methadone-metabolizing enzyme, and by a locus 300 kb 5" to OPRM1. Methadone 121-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 28639119-1 2017 PURPOSE: In vitro data showed that selexipag and its active metabolite (ACT-333679) have an inductive effect on CYP3A4, CYP2B6, and CYP2C9 at concentrations approximately 100-fold higher than the maximum plasma concentration (C max) measured under steady-state conditions. selexipag 35-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 28187506-7 2017 Nevirapine group: CYP2B6 516G>T was associated with higher log10 Cmax and lower log10 Cmin . Nevirapine 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 28819225-5 2017 In contrast, P450/ABC transporter induction with rifampicin markedly impaired ivermectin absorption. Rifampin 49-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 28718515-2 2017 Although primarily metabolized by CYP2B6 and -3A, efavirenz (EFV) and lopinavir/ritonavir (LPV/r) are substrates of P-glycoprotein and the solute carrier organic (SLCO) anion transporter, respectively. Lopinavir 70-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 28718515-2 2017 Although primarily metabolized by CYP2B6 and -3A, efavirenz (EFV) and lopinavir/ritonavir (LPV/r) are substrates of P-glycoprotein and the solute carrier organic (SLCO) anion transporter, respectively. Ritonavir 80-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 28718515-8 2017 CYP2B6 516G T was significantly associated with EFV concentrations (p<0.001). efavirenz 48-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 28559276-9 2017 CYP2B6*6 genotype polymorphisms were associated with decreased EFV and 8OHEFV clearance. efavirenz 63-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 28481785-2 2017 Our objective was to investigate the relationship between the CYP2B6 516G>T genotype and late virologic failure in patients treated with EFV in Gaborone, Botswana. efavirenz 140-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 28481785-2 2017 Our objective was to investigate the relationship between the CYP2B6 516G>T genotype and late virologic failure in patients treated with EFV in Gaborone, Botswana. gaborone 147-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 28481785-10 2017 CONCLUSION: The CYP2B6 516 T-allele was protective against late virologic breakthrough in patients with initial (6 month) HIV RNA suppression on EFV-based ART. efavirenz 145-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 27853934-8 2017 RESULTS: N-oxides of clozapine (CYP2B6/2C19) and voriconazole (CYP2C9/3A4) showed CYP inhibition >=50%. n-oxides 9-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 27853934-8 2017 RESULTS: N-oxides of clozapine (CYP2B6/2C19) and voriconazole (CYP2C9/3A4) showed CYP inhibition >=50%. Clozapine 21-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 27853934-9 2017 Clozapine-N-oxide inhibited CYP2B6 and CYP2C19 pathways with IC50 of 8.3 and 8.7 microM, respectively. clozapine N-oxide 0-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 27853934-10 2017 Voriconazole-N-oxide inhibited CYP2B6 and CYP2C19 pathways with IC50 of 10.5 and 11.2 microM, respectively. Voriconazole N-Oxide 0-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 27853934-11 2017 Co-incubation of clozapine-N-oxide with clozapine potentiated CYP2B6/2C19 pathways; however, incubation of voriconazole-N-oxide with voriconazole did not appear to potentiate the CYP pathways because parent caused an inhibition of almost 80%. clozapine N-oxide 17-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 27853934-11 2017 Co-incubation of clozapine-N-oxide with clozapine potentiated CYP2B6/2C19 pathways; however, incubation of voriconazole-N-oxide with voriconazole did not appear to potentiate the CYP pathways because parent caused an inhibition of almost 80%. Clozapine 17-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 28559276-9 2017 CYP2B6*6 genotype polymorphisms were associated with decreased EFV and 8OHEFV clearance. 8ohefv 71-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 28340497-0 2017 P450 inhibitor ketoconazole increased the intratumor drug levels and antitumor activity of fenretinide in human neuroblastoma xenograft models. Ketoconazole 15-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 28724407-2 2017 These enzymes require electrons for their activity, and the electrons are supplied by NAD(P)H through a P450 electron donor system, which is generally a cytochrome P450 reductase (CPR). nad(p)h 86-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-108 28724407-3 2017 The yeast Xanthophyllomyces dendrorhous has evolved an exclusive P450-CPR system that specializes in the synthesis of astaxanthin, a carotenoid with commercial potential. astaxanthine 118-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 28724407-3 2017 The yeast Xanthophyllomyces dendrorhous has evolved an exclusive P450-CPR system that specializes in the synthesis of astaxanthin, a carotenoid with commercial potential. Carotenoids 133-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 28724407-6 2017 Structural analyses of the X. dendrorhous P450 proteins showed that all of them have a predicted transmembrane region at their N-terminus and have the conserved domains characteristic of the P450s, including the heme-binding region (FxxGxRxCxG); the PER domain, with the characteristic signature for fungi (PxRW); the ExxR motif in the K-helix region and the oxygen-binding domain (OBD) (AGxDTT); also, the characteristic secondary structure elements of all the P450 proteins were identified. Heme 212-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 28724407-6 2017 Structural analyses of the X. dendrorhous P450 proteins showed that all of them have a predicted transmembrane region at their N-terminus and have the conserved domains characteristic of the P450s, including the heme-binding region (FxxGxRxCxG); the PER domain, with the characteristic signature for fungi (PxRW); the ExxR motif in the K-helix region and the oxygen-binding domain (OBD) (AGxDTT); also, the characteristic secondary structure elements of all the P450 proteins were identified. Heme 212-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 191-195 28724407-6 2017 Structural analyses of the X. dendrorhous P450 proteins showed that all of them have a predicted transmembrane region at their N-terminus and have the conserved domains characteristic of the P450s, including the heme-binding region (FxxGxRxCxG); the PER domain, with the characteristic signature for fungi (PxRW); the ExxR motif in the K-helix region and the oxygen-binding domain (OBD) (AGxDTT); also, the characteristic secondary structure elements of all the P450 proteins were identified. Oxygen 359-365 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 28724407-6 2017 Structural analyses of the X. dendrorhous P450 proteins showed that all of them have a predicted transmembrane region at their N-terminus and have the conserved domains characteristic of the P450s, including the heme-binding region (FxxGxRxCxG); the PER domain, with the characteristic signature for fungi (PxRW); the ExxR motif in the K-helix region and the oxygen-binding domain (OBD) (AGxDTT); also, the characteristic secondary structure elements of all the P450 proteins were identified. Oxygen 359-365 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 191-195 28340497-0 2017 P450 inhibitor ketoconazole increased the intratumor drug levels and antitumor activity of fenretinide in human neuroblastoma xenograft models. Fenretinide 91-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 28340497-1 2017 We previously reported that concurrent ketoconazole, an oral anti-fungal agent and P450 enzyme inhibitor, increased plasma levels of the cytotoxic retinoid, fenretinide (4-HPR) in mice. Ketoconazole 39-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-87 28340497-1 2017 We previously reported that concurrent ketoconazole, an oral anti-fungal agent and P450 enzyme inhibitor, increased plasma levels of the cytotoxic retinoid, fenretinide (4-HPR) in mice. Retinoids 147-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-87 28340497-1 2017 We previously reported that concurrent ketoconazole, an oral anti-fungal agent and P450 enzyme inhibitor, increased plasma levels of the cytotoxic retinoid, fenretinide (4-HPR) in mice. Fenretinide 157-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-87 28320034-0 2017 Variations in Infant CYP2B6 Genotype Associated with the Need for Pharmacological Treatment for Neonatal Abstinence Syndrome in Infants of Methadone-Maintained Opioid-Dependent Mothers. Methadone 139-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 28403028-4 2017 RECENT FINDINGS: Dose adjustments of standard dose efavirenz are not necessary with rifampicin, because auto-induction of CYP2B6 by efavirenz counteracts the induction of rifampicin. efavirenz 132-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 29060457-2 2017 Clopidogrel is metabolized by hepatic CYP2C19 and CYP2B6, therefore, co-administration of clopidogrel and CYP2C19 inhibitors can alter pharmacokinetics of clopidogrel. Clopidogrel 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 29060457-2 2017 Clopidogrel is metabolized by hepatic CYP2C19 and CYP2B6, therefore, co-administration of clopidogrel and CYP2C19 inhibitors can alter pharmacokinetics of clopidogrel. Clopidogrel 90-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 29060457-2 2017 Clopidogrel is metabolized by hepatic CYP2C19 and CYP2B6, therefore, co-administration of clopidogrel and CYP2C19 inhibitors can alter pharmacokinetics of clopidogrel. Clopidogrel 155-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 28403028-4 2017 RECENT FINDINGS: Dose adjustments of standard dose efavirenz are not necessary with rifampicin, because auto-induction of CYP2B6 by efavirenz counteracts the induction of rifampicin. Rifampin 171-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 28403028-5 2017 However, patients with extensive metabolizer CYP2B6 genotypes have lower efavirenz concentrations and may have less auto-induction of CYP2B6; the additive inducing effects of rifampicin on CYP2B6 could result in clinically significant reductions of efavirenz concentrations. Rifampin 175-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 28427998-4 2017 Treatment of the cells with the NO donor (Z)-1-[N-(3-aminopropyl)-N-(3-ammoniopropyl)amino]diazen-1-ium-1,2-diolate also resulted in rapid down-regulation of CYP2B6 activity, measured as the formation of 7-hydroxy-4-trifluoromethylcoumarin, as well as 2B6 protein in the CYP2B6 HeLa cell line. SCHEMBL17102363 41-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 271-277 28427998-0 2017 Nitric oxide-regulated proteolysis of human CYP2B6 via the ubiquitin-proteasome system. Nitric Oxide 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 28427998-8 2017 The cellular NO produced by doxycycline treatment also effectively down-regulated CYP2B6 protein, which was blocked by the co-treatment with the NOS2 competitive inhibitor L-NG-nitroarginine methyl ester (L-NAME). Doxycycline 28-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 28427998-3 2017 Native CYP2B6 protein was rapidly down-regulated in HeLa cells within 3h of treatment with the NO donor (Z)-1-[2-(2-Aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate, while its mRNA level was not down-regulated. SCHEMBL1617643 104-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 7-13 28427998-8 2017 The cellular NO produced by doxycycline treatment also effectively down-regulated CYP2B6 protein, which was blocked by the co-treatment with the NOS2 competitive inhibitor L-NG-nitroarginine methyl ester (L-NAME). NG-Nitroarginine Methyl Ester 172-203 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 28427998-4 2017 Treatment of the cells with the NO donor (Z)-1-[N-(3-aminopropyl)-N-(3-ammoniopropyl)amino]diazen-1-ium-1,2-diolate also resulted in rapid down-regulation of CYP2B6 activity, measured as the formation of 7-hydroxy-4-trifluoromethylcoumarin, as well as 2B6 protein in the CYP2B6 HeLa cell line. SCHEMBL17102363 41-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-164 28427998-8 2017 The cellular NO produced by doxycycline treatment also effectively down-regulated CYP2B6 protein, which was blocked by the co-treatment with the NOS2 competitive inhibitor L-NG-nitroarginine methyl ester (L-NAME). NG-Nitroarginine Methyl Ester 205-211 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 28427998-13 2017 Further investigation demonstrated that CYP2B6 protein was polyubiquitinated and this was dramatically enhanced by co-treatment with NO donor and bortezomib. Bortezomib 146-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 28384415-1 2017 The marked induction of cytochromes P450 such as CYP1A1 caused by polycyclic aromatic hydrocarbons (PAHs) like 3-methylcholanthrene (MC) is often accompanied by suppression of other hepatic P450s. Polycyclic Aromatic Hydrocarbons 66-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 28181240-5 2017 Samples with CYP2B6*6 and/or CYP2C19*2 had lower exposure to 4-hydroxycyclophosphamide, whereas those with CYP2B6*1G/*1H had higher exposure. 4-hydroxycyclophosphamide 61-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 28181240-11 2017 Genetic variations in CYP2B6, CYP2C19, and CYP3A5 dramatically affected the mRNA expression of proteins, the pharmacokinetics of 4-hydroxycyclophosphamide, and the R-CHOP treatment response in Chinese subjects. 4-hydroxycyclophosphamide 129-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 28288493-4 2017 Furthermore, the inhibition was potentiated following preincubation with NADPH, indicating that G. cambogia extract is a time-dependent inhibitor of CYP2B6. NADP 73-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 28611384-7 2017 Compared to the high tacrolimus group, expression levels of six cytochrome P450 enzymes, CYP1A1, CYP2B6, CYP3A5, CYP4A11, CYP19A1, and CYP17A1 were significantly higher in the low tacrolimus group. Tacrolimus 180-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 28858450-4 2017 A small body of literature indicates that drugs that induce cytochrome P450 (CYP)2B6 and CYP3A4 will reduce exposure to ketamine and that drugs that inhibit these enzymes will increase exposure to ketamine. Ketamine 120-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-84 28858450-5 2017 Common genetic polymorphisms of the CYP2B6 gene may also be associated with variations in the exposure to ketamine. Ketamine 106-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 27832320-10 2017 Finally, in human hepatocyte-like HepaRG cells, PFOA treatment increased mRNA levels of CYP2B6, a CAR target gene, as did phenobarbital. perfluorooctanoic acid 48-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 28384415-1 2017 The marked induction of cytochromes P450 such as CYP1A1 caused by polycyclic aromatic hydrocarbons (PAHs) like 3-methylcholanthrene (MC) is often accompanied by suppression of other hepatic P450s. Polycyclic Aromatic Hydrocarbons 100-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 27896399-5 2017 Seven isoforms were capable of metabolizing triclosan, with the order of activity being CYP1A2 > CYP2B6 > CYP2C19 > CYP2D6 CYP1B1 > CYP2C18 CYP1A1. Triclosan 44-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 28384415-1 2017 The marked induction of cytochromes P450 such as CYP1A1 caused by polycyclic aromatic hydrocarbons (PAHs) like 3-methylcholanthrene (MC) is often accompanied by suppression of other hepatic P450s. Methylcholanthrene 111-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 28384415-1 2017 The marked induction of cytochromes P450 such as CYP1A1 caused by polycyclic aromatic hydrocarbons (PAHs) like 3-methylcholanthrene (MC) is often accompanied by suppression of other hepatic P450s. Methylcholanthrene 133-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 28384415-3 2017 MC suppresses mRNA levels for CYP2C8, an important human P450, in cultured human hepatocytes. Methylcholanthrene 0-2 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 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 2 subfamily B member 6 Homo sapiens 121-127 28283499-6 2017 CYP2B6 and to an even lesser extent CYP2C9 were also observed to catalyze NGMN metabolism. ngmn 74-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 28368100-0 2017 Halogen-pi Interactions in the Cytochrome P450 Active Site: Structural Insights into Human CYP2B6 Substrate Selectivity. Halogens 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 28369648-4 2017 P450 enzymes metabolize chlorpyrifos to chlorpyrifos-oxon, which is then metabolized primarily to 3, 5, 6-trichloropyridinol in addition to diethylphosphate and diethylthiophosphate. Chlorpyrifos 24-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 28369648-4 2017 P450 enzymes metabolize chlorpyrifos to chlorpyrifos-oxon, which is then metabolized primarily to 3, 5, 6-trichloropyridinol in addition to diethylphosphate and diethylthiophosphate. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 40-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 28369648-4 2017 P450 enzymes metabolize chlorpyrifos to chlorpyrifos-oxon, which is then metabolized primarily to 3, 5, 6-trichloropyridinol in addition to diethylphosphate and diethylthiophosphate. 3, 5, 6-trichloropyridinol 98-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 28369648-4 2017 P450 enzymes metabolize chlorpyrifos to chlorpyrifos-oxon, which is then metabolized primarily to 3, 5, 6-trichloropyridinol in addition to diethylphosphate and diethylthiophosphate. diethyl phosphate 140-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 28369648-4 2017 P450 enzymes metabolize chlorpyrifos to chlorpyrifos-oxon, which is then metabolized primarily to 3, 5, 6-trichloropyridinol in addition to diethylphosphate and diethylthiophosphate. thiophosphoric acid 161-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 28369648-9 2017 The P450 inhibitor SKF525A partly negated the protective effect of astrocytes, allowing reduction in branch points with chlorpyrifos (10 muM). Proadifen 19-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-8 28369648-9 2017 The P450 inhibitor SKF525A partly negated the protective effect of astrocytes, allowing reduction in branch points with chlorpyrifos (10 muM). Chlorpyrifos 120-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-8 28369648-10 2017 Thus, the scalable and defined astrocyte-neuron cocultures model that we established here has potentially identified a role for P450 enzymes in astrocytic neuroprotection against chlorpyrifos and provides a novel model for addressing DNT in a more accurate multicellular environment. Chlorpyrifos 179-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 28368100-2 2017 To assess the role of halogen-pi bonds in substrate selectivity and orientation in the active site, structures of four CYP2B6 monoterpenoid complexes were solved by X-ray crystallography. Halogens 22-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 28368100-2 2017 To assess the role of halogen-pi bonds in substrate selectivity and orientation in the active site, structures of four CYP2B6 monoterpenoid complexes were solved by X-ray crystallography. Monoterpenes 126-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 28368100-5 2017 The bromine in myrtenyl bromide pi-bonded with Phe297 in CYP2B6, whereas the two major orientations in the active site mutant I114V exhibited bromine-pi interactions with two additional residues, Phe108 and Phe115. Bromine 4-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 28368100-5 2017 The bromine in myrtenyl bromide pi-bonded with Phe297 in CYP2B6, whereas the two major orientations in the active site mutant I114V exhibited bromine-pi interactions with two additional residues, Phe108 and Phe115. myrtenyl bromide 15-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 28368100-6 2017 Analysis of existing structures suggests that halogen-pi interactions may be unique to the CYP2B enzymes within CYP family 2 but are also important for CYP3A enzymes. Halogens 46-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-96 28468305-4 2017 Fargesin negligibly inhibited CYP1A2-catalyzed phenacetin O-deethylation, CYP2A6-catalyzed coumarin 7-hydroxylation, CYP2B6-catalyzed bupropion hydroxylation, and CYP2D6-catalyzed bufuralol 1"-hydroxylation at 100 muM in human liver microsomes. Bupropion 134-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 28184434-0 2017 Tell-Tale SNPs: The Role of CYP2B6 in Methadone Fatalities. Methadone 38-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 28184434-4 2017 The purpose of this study was to determine if one or more single nucleotide polymorphisms (SNPs) located within the CYP2B6 gene contributes to a poor metabolizer phenotype for methadone in these fatal cases. Methadone 176-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 28184434-9 2017 These results indicate that these three CYP2B6 SNPs are associated with methadone fatalities. Methadone 72-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 28342136-1 2017 Cytochrome P450 OleTJE has attracted much attention for its ability to catalyze the decarboxylation of long chain fatty acids to generate alkenes, which are not only biofuel molecule, but also can be used broadly for making lubricants, polymers and detergents. long chain fatty acids 103-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 28342136-1 2017 Cytochrome P450 OleTJE has attracted much attention for its ability to catalyze the decarboxylation of long chain fatty acids to generate alkenes, which are not only biofuel molecule, but also can be used broadly for making lubricants, polymers and detergents. Alkenes 138-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 28342136-1 2017 Cytochrome P450 OleTJE has attracted much attention for its ability to catalyze the decarboxylation of long chain fatty acids to generate alkenes, which are not only biofuel molecule, but also can be used broadly for making lubricants, polymers and detergents. Polymers 236-244 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 28342136-2 2017 In this study, the molecular basis of the binding mechanism of P450 OleTJE for arachidic acid, myristic acid, and caprylic acid was investigated by utilizing conventional molecular dynamics simulation and binding free energy calculations. arachidic acid 79-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 28342136-2 2017 In this study, the molecular basis of the binding mechanism of P450 OleTJE for arachidic acid, myristic acid, and caprylic acid was investigated by utilizing conventional molecular dynamics simulation and binding free energy calculations. Myristic Acid 95-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 28342136-2 2017 In this study, the molecular basis of the binding mechanism of P450 OleTJE for arachidic acid, myristic acid, and caprylic acid was investigated by utilizing conventional molecular dynamics simulation and binding free energy calculations. octanoic acid 114-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 28342136-7 2017 It is suggested that the reaction is easier to carry out in myristic acid bound system than those in arachidic acid and caprylic acid bound system based on the distance of Hbeta atom of substrate relative to P450 OleTJE Compound I states. Myristic Acid 60-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 208-212 28342136-7 2017 It is suggested that the reaction is easier to carry out in myristic acid bound system than those in arachidic acid and caprylic acid bound system based on the distance of Hbeta atom of substrate relative to P450 OleTJE Compound I states. arachidic acid 101-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 208-212 28342136-7 2017 It is suggested that the reaction is easier to carry out in myristic acid bound system than those in arachidic acid and caprylic acid bound system based on the distance of Hbeta atom of substrate relative to P450 OleTJE Compound I states. octanoic acid 120-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 208-212 28342136-8 2017 This study provided novel insight to understand the substrate preference mechanism of P450 OleTJE and valuable information for rational enzyme design for short chain fatty acid decarboxylation. Fatty Acids, Volatile 154-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-90 27826892-12 2017 From all the analysed changes, only polymorphism c.516G>T in the CYP2B6 gene and BMI affect the metabolism rate of propofol and may play an important role in the optimisation of propofol anaesthesia. Propofol 118-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 27826892-12 2017 From all the analysed changes, only polymorphism c.516G>T in the CYP2B6 gene and BMI affect the metabolism rate of propofol and may play an important role in the optimisation of propofol anaesthesia. Propofol 181-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 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 2 subfamily B member 6 Homo sapiens 71-77 28188297-1 2017 The human genome encodes 57 cytochrome P450 genes, whose enzyme products metabolize hundreds of drugs, thousands of xenobiotics, and unknown numbers of endogenous compounds, including steroids, retinoids, and eicosanoids. Steroids 184-192 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 28235828-5 2017 Genome-wide association analyses identified significant maternal loci for p,p"-DDE (P = 7.8 x 10-11) in the CYP2B6 gene and for BDE-28 (P = 3.2 x 10-8) near the SH3GL2 gene, both involved in xenobiotic and lipid metabolism. Dichlorodiphenyl Dichloroethylene 74-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 27861737-4 2017 As for carbon monoxide difference absorbance spectroscopy, used for concentration measurements of active P450 monooxygenases (P450s), security standards, and consistent gassing have to be addressed. Carbon Monoxide 7-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-109 27861737-8 2017 The designed method was successfully applied to concentration measurements and carbon monoxide (CO) saturation kinetics ranging from 0.3 to 5.0 muM P450 BM3 achieving a measurement accuracy of +-0.05 muM P450. Carbon Monoxide 79-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 27861737-8 2017 The designed method was successfully applied to concentration measurements and carbon monoxide (CO) saturation kinetics ranging from 0.3 to 5.0 muM P450 BM3 achieving a measurement accuracy of +-0.05 muM P450. Carbon Monoxide 79-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 204-208 27861737-8 2017 The designed method was successfully applied to concentration measurements and carbon monoxide (CO) saturation kinetics ranging from 0.3 to 5.0 muM P450 BM3 achieving a measurement accuracy of +-0.05 muM P450. Carbon Monoxide 96-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 27861737-8 2017 The designed method was successfully applied to concentration measurements and carbon monoxide (CO) saturation kinetics ranging from 0.3 to 5.0 muM P450 BM3 achieving a measurement accuracy of +-0.05 muM P450. Carbon Monoxide 96-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 204-208 28520385-3 2012 Prasugrel is metabolized to its active metabolite primarily by CYP3A5 and CYP2B6, and to a lesser extent by CYP2C9 and CYP2C19. Prasugrel Hydrochloride 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 28188297-1 2017 The human genome encodes 57 cytochrome P450 genes, whose enzyme products metabolize hundreds of drugs, thousands of xenobiotics, and unknown numbers of endogenous compounds, including steroids, retinoids, and eicosanoids. Retinoids 194-203 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 28188297-1 2017 The human genome encodes 57 cytochrome P450 genes, whose enzyme products metabolize hundreds of drugs, thousands of xenobiotics, and unknown numbers of endogenous compounds, including steroids, retinoids, and eicosanoids. Eicosanoids 209-220 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 28252572-2 2017 OBJECTIVES: The aim of the study was to investigate the feasibility and sample sizes required to explore the relationship between CYP2B6*6 and GRIN2B single-nucleotide polymorphisms and ketamine-induced EP. Ketamine 186-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 28179134-3 2017 The results demonstrate that most of the unsaturated fatty acids showed marked inhibition towards CYP2C8 mediated amodiaquine N-deethylation followed by inhibition of CYP2C9 and CYP2B6 mediated activities. Fatty Acids, Unsaturated 41-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-184 28263461-8 2017 However, CYP2B6 is also a major catalyst of methadone metabolism in vitro. Methadone 44-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 28263461-9 2017 It has now been unequivocally established that CYP2B6, not CYP3A4, is the principal determinant of methadone metabolism, clearance, elimination, and plasma concentrations in humans. Methadone 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 28263461-11 2017 CYP2B6 genetics also influences methadone metabolism and clearance, which were diminished in CYP2B6*6 carriers and increased in CYP2B6*4 carriers. Methadone 32-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 28263461-11 2017 CYP2B6 genetics also influences methadone metabolism and clearance, which were diminished in CYP2B6*6 carriers and increased in CYP2B6*4 carriers. Methadone 32-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 28263461-11 2017 CYP2B6 genetics also influences methadone metabolism and clearance, which were diminished in CYP2B6*6 carriers and increased in CYP2B6*4 carriers. Methadone 32-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 28263461-12 2017 CYP2B6 genetics can explain, in part, interindividual variability in methadone metabolism and clearance. Methadone 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 28263461-13 2017 Thus, both constitutive variability due to CYP2B6 genetics, and CYP2B6-mediated drug interactions, can alter methadone disposition, clinical effect, and drug safety. Methadone 109-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 28263461-13 2017 Thus, both constitutive variability due to CYP2B6 genetics, and CYP2B6-mediated drug interactions, can alter methadone disposition, clinical effect, and drug safety. Methadone 109-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 28117133-0 2017 Pharmacogenetics of ecstasy: CYP1A2, CYP2C19, and CYP2B6 polymorphisms moderate pharmacokinetics of MDMA in healthy subjects. N-Methyl-3,4-methylenedioxyamphetamine 100-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 28117133-1 2017 In vitro studies showed that CYP2C19, CYP2B6, and CYP1A2 contribute to the metabolism of 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) to 3,4-methylenedioxyamphetamine (MDA). N-Methyl-3,4-methylenedioxyamphetamine 89-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 28117133-1 2017 In vitro studies showed that CYP2C19, CYP2B6, and CYP1A2 contribute to the metabolism of 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) to 3,4-methylenedioxyamphetamine (MDA). N-Methyl-3,4-methylenedioxyamphetamine 124-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 28117133-1 2017 In vitro studies showed that CYP2C19, CYP2B6, and CYP1A2 contribute to the metabolism of 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) to 3,4-methylenedioxyamphetamine (MDA). 3,4-Methylenedioxyamphetamine 142-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 28117133-1 2017 In vitro studies showed that CYP2C19, CYP2B6, and CYP1A2 contribute to the metabolism of 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) to 3,4-methylenedioxyamphetamine (MDA). 3,4-Methylenedioxyamphetamine 173-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 28117133-7 2017 The findings indicate that CYP2C19, CYP2B6, and CYP1A2 contribute to the metabolism of MDMA to MDA in humans. N-Methyl-3,4-methylenedioxyamphetamine 87-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 27815210-1 2017 Steroids are synthesized from the adrenal glands and gonads by enzymes of the cytochromes P450 and hydroxysteroid dehydrogenase in nature. Steroids 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-127 27574897-2 2017 beta-Ionone was found to be oxidized via 4S-hydroxylation by CYP2B6 in human liver microsomes. beta-ionone 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 26956662-11 2017 Feline CYP2B6 proteins heterologously expressed in Escherichia coli metabolized several substrates specific to human CYP2B6, including 7-ethoxy-4-(trifluoromethyl) coumarin (EFC). 7-ethoxy-4-trifluoromethylcoumarin 135-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 7-13 26956662-11 2017 Feline CYP2B6 proteins heterologously expressed in Escherichia coli metabolized several substrates specific to human CYP2B6, including 7-ethoxy-4-(trifluoromethyl) coumarin (EFC). 7-ethoxy-4-trifluoromethylcoumarin 135-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 26956662-11 2017 Feline CYP2B6 proteins heterologously expressed in Escherichia coli metabolized several substrates specific to human CYP2B6, including 7-ethoxy-4-(trifluoromethyl) coumarin (EFC). 7-ethoxy-4-trifluoromethylcoumarin 174-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 7-13 26956662-11 2017 Feline CYP2B6 proteins heterologously expressed in Escherichia coli metabolized several substrates specific to human CYP2B6, including 7-ethoxy-4-(trifluoromethyl) coumarin (EFC). 7-ethoxy-4-trifluoromethylcoumarin 174-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 27574897-7 2017 These results suggest that beta-ionone, a norisoprenoid present in nature, is one of the effective substrates for CYP2B enzymes in human liver microsomes. beta-ionone 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-119 27574897-2 2017 beta-Ionone was found to be oxidized via 4S-hydroxylation by CYP2B6 in human liver microsomes. 4s 41-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 27574897-7 2017 These results suggest that beta-ionone, a norisoprenoid present in nature, is one of the effective substrates for CYP2B enzymes in human liver microsomes. Norisoprenoids 42-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-119 26956662-12 2017 The metabolic activity was strongly inhibited by medetomidine and atipamezole, potent inhibitors of canine CYP2B11 (now officially CYP2B6) as well as by ticlopidine and sertraline, inhibitors selective to human CYP2B6. Medetomidine 49-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 211-217 26956662-12 2017 The metabolic activity was strongly inhibited by medetomidine and atipamezole, potent inhibitors of canine CYP2B11 (now officially CYP2B6) as well as by ticlopidine and sertraline, inhibitors selective to human CYP2B6. atipamezole 66-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 211-217 26956662-12 2017 The metabolic activity was strongly inhibited by medetomidine and atipamezole, potent inhibitors of canine CYP2B11 (now officially CYP2B6) as well as by ticlopidine and sertraline, inhibitors selective to human CYP2B6. Sertraline 169-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 211-217 27574897-4 2017 Further kinetic analysis revealed that CYP2B6 exhibited the highest activity for beta-ionone 4-hydroxylation. beta-ionone 81-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 27574897-5 2017 Kinetic analysis showed that Km and Vmax for oxidation of beta-ionone by CYP1A2 and CYP2B6 was 107.9 +- 36.0 microM and 3200.3 +- 323.0 nmol/min/nmol P450 and 5.6 +- 1.2 microM and 572.8 +- 29.8 nmol/min/nmol P450, respectively. beta-ionone 58-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 27750412-2 2017 Thalidomide exerts its various pharmacological and toxic actions in primates through multiple mechanisms, including nonspecific modification of many protein networks after bioactivation by autoinduced human P450 enzymes. Thalidomide 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 207-211 27750412-6 2017 Genome-wide association studies in humans have been rapidly advanced; however, unique whole-gene deletion of P450 2A6 was subsequently developed to cover nicotine-related lung cancer risk study. Nicotine 154-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 27707991-0 2017 Effect of diurnal variation, CYP2B6 genotype and age on the pharmacokinetics of nevirapine in African children. Nevirapine 80-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 27871908-5 2017 We demonstrate that the DCA 3,12-diacetate and CA 3,7,12-triacetate derivatives are ligands of PXR and DCA 3,12-diacetate induces PXR target genes such as CYP3A4, CYP2B6 and ABCB1/MDR1. dca 3,12-diacetate 24-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 27871908-5 2017 We demonstrate that the DCA 3,12-diacetate and CA 3,7,12-triacetate derivatives are ligands of PXR and DCA 3,12-diacetate induces PXR target genes such as CYP3A4, CYP2B6 and ABCB1/MDR1. ca 3,7,12-triacetate 47-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 28154789-0 2017 CYP2B6 Genotype Guided Dosing of Propofol Anesthesia in the Elderly based on Nonparametric Population Pharmacokinetic Modeling and Simulations. Propofol 33-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 28154789-10 2017 Model-based dosing-simulations comparing patients with the CYP2B6 AA & AG genotypes to both GG genotypes and patients from a multicenter trial suggest a 50% decrease in propofol infusion dose, to 25mg/kg/min, be made to result in approximately equivalent drug exposures. Adenosine Monophosphate 70-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 28154789-10 2017 Model-based dosing-simulations comparing patients with the CYP2B6 AA & AG genotypes to both GG genotypes and patients from a multicenter trial suggest a 50% decrease in propofol infusion dose, to 25mg/kg/min, be made to result in approximately equivalent drug exposures. Propofol 173-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 28154789-11 2017 CONCLUSION: Based on the pharmacometric modeling and simulation, if no dosage adjustments are made for the elderly CYP2B6 AA and AG genotypes, a 250% higher propofol blood exposure will be evident within 1-hour from the start of the infusion. Propofol 157-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 28154789-12 2017 Thus, based on the pharmacokinetic model, genotyping elderly patients for the CYP2B6 AA and AG gene variants will decrease the total propofol blood exposure during anesthesia and sedation when an infusion dose adjustment is made to 25mg/kg/min. Propofol 133-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 27871908-5 2017 We demonstrate that the DCA 3,12-diacetate and CA 3,7,12-triacetate derivatives are ligands of PXR and DCA 3,12-diacetate induces PXR target genes such as CYP3A4, CYP2B6 and ABCB1/MDR1. dca 3,12-diacetate 103-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 27273149-4 2017 Thus, isavuconazole is a mild inducer of CYP2B6 but does not appear to affect CYP1A2-, CYP2C8-, or CYP2D6-mediated metabolism. isavuconazole 6-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 27839851-0 2017 Genetic and methylation variation in the CYP2B6 gene is related to circulating p,p"-dde levels in a population-based sample. Dichlorodiphenyl Dichloroethylene 79-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 27839851-6 2017 RESULTS: Evidence for genome-wide significant association with p,p"-DDE levels was observed only for a locus at chromosome 19 corresponding to the CYP2B6 gene (lead SNP rs7260538). Dichlorodiphenyl Dichloroethylene 63-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 27839851-9 2017 A whole-genome methylation analysis showed one significant relationship vs. p,p"-DDE levels (p=6.2x10-9) located 7kb downstream the CYP2B6 gene (cg27089200, position chr19:41531976). Dichlorodiphenyl Dichloroethylene 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-138 27839851-11 2017 CONCLUSION: Circulating levels of p,p"-DDE were related to genetic variation in the CYP2B6 gene in the general elderly population. Dichlorodiphenyl Dichloroethylene 34-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 29081796-2 2017 Examples of CYP2B6"s impacts include its linkage to mortality during cyclophosphamide therapy and its role in determining hepatotoxicity and CNS toxicity during efavirenz therapy for HIV infection. Cyclophosphamide 69-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 27799204-8 2017 Apparent oral clearance of efavirenz was reduced by 25% and 51% in subjects predicted to have intermediate and slow CYP2B6 metabolizer status, respectively. efavirenz 27-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 29142176-8 2017 NaPB and momfluorothrin also increased CYP2B6 mRNA levels in human hepatocytes. momfluorothrin 9-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 28006013-6 2016 Aflatoxin B1 and benzo(a)pyrene were predominantly detected by the CYP3A4 + RAD54 system, while N-nitrosodimethylamine only moderately activated the CYP2B6 + RAD54 reporter system and none of them was identified by the CYP2D6 + RAD54 system. Dimethylnitrosamine 96-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 28081342-2 2016 Nevirapine is extensively metabolized in the liver and CYP2B6 is mainly responsible for oxidation of 3-hydroxynevirapine (3-OH NVP). 3-Hydroxy Nevirapine 101-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 28081342-3 2016 This study aims to explore CYP2B6 activity by measuring 2-hydroxynevirapine (2-OH NVP) and 3-OH NVP in plasma and to identify factors associated with nevirapine pharmacokinetic parameters. 2-Hydroxy Nevirapine 56-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 27860311-0 2016 Single Heteroatom Substitutions in the Efavirenz Oxazinone Ring Impact Metabolism by CYP2B6. efavirenz oxazinone 39-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 27860311-1 2016 Previously, we observed that the oxazinone ring is important for cytochrome P450 2B6 (CYP2B6) activity toward efavirenz ((4S)-6-chloro-4-(2-cyclopropylethynyl)-1,4-dihydro-4-(trifluoromethyl)-2H-3,1-benzoxazin-2-one), a CYP2B6 substrate used to treat HIV. oxazinone 33-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-84 27860311-1 2016 Previously, we observed that the oxazinone ring is important for cytochrome P450 2B6 (CYP2B6) activity toward efavirenz ((4S)-6-chloro-4-(2-cyclopropylethynyl)-1,4-dihydro-4-(trifluoromethyl)-2H-3,1-benzoxazin-2-one), a CYP2B6 substrate used to treat HIV. oxazinone 33-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 27860311-1 2016 Previously, we observed that the oxazinone ring is important for cytochrome P450 2B6 (CYP2B6) activity toward efavirenz ((4S)-6-chloro-4-(2-cyclopropylethynyl)-1,4-dihydro-4-(trifluoromethyl)-2H-3,1-benzoxazin-2-one), a CYP2B6 substrate used to treat HIV. oxazinone 33-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-226 27860311-7 2016 Our work reveals that analogues with heteroatom changes in the oxazinone ring are still CYP2B6 substrates, although the changes in KM suggest altered substrate binding. oxazinone 63-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 27763886-0 2016 CYP2B6*6 or Not CYP2B6*6-That Remains a Question for Precision Medicine and Ketamine! Ketamine 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27763887-13 2016 CONCLUSIONS: These results show that while the CYP2B6*6 polymorphism results in diminished ketamine metabolism in vitro, this allelic variant did not affect single, low-dose ketamine metabolism, clearance, and pharmacokinetics in vivo. Ketamine 91-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 27763886-0 2016 CYP2B6*6 or Not CYP2B6*6-That Remains a Question for Precision Medicine and Ketamine! Ketamine 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 27763887-1 2016 BACKGROUND: At therapeutic concentrations, cytochrome P4502B6 (CYP2B6) is the major P450 isoform catalyzing hepatic ketamine N-demethylation to norketamine in vitro. Ketamine 116-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 27763887-1 2016 BACKGROUND: At therapeutic concentrations, cytochrome P4502B6 (CYP2B6) is the major P450 isoform catalyzing hepatic ketamine N-demethylation to norketamine in vitro. norketamine 144-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 27763887-4 2016 CYP2B6.6, the protein encoded by the CYP2B6*6 allele, and liver microsomes from CYP2B6*6 carriers had diminished ketamine metabolism in vitro. Ketamine 113-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27763887-4 2016 CYP2B6.6, the protein encoded by the CYP2B6*6 allele, and liver microsomes from CYP2B6*6 carriers had diminished ketamine metabolism in vitro. Ketamine 113-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 27763887-4 2016 CYP2B6.6, the protein encoded by the CYP2B6*6 allele, and liver microsomes from CYP2B6*6 carriers had diminished ketamine metabolism in vitro. Ketamine 113-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 27763887-5 2016 This investigation tested whether humans with the CYP2B6*6 allele would have decreased clinical ketamine metabolism and clearance. Ketamine 96-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 27917125-3 2016 Under non-induced conditions, all CYP activities could be determined in 3D-PHH, CYP2B6, CYP2C19, CYP2D6, and CYP3A4 in 2D-PHH and HepaRG, and CYP2C19 and CYP3A4 in HepG2 cells. heparg 130-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 27286724-8 2016 (S)-methadone clearance was influenced by cytochrome P450 (CYP) 2B6 activity, ABCB1 3435C>T, and alpha-1 acid glycoprotein level, while (R)-methadone clearance was influenced by CYP2B6 activity, POR*28, and CYP3A4*22. Methadone 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-67 27286724-8 2016 (S)-methadone clearance was influenced by cytochrome P450 (CYP) 2B6 activity, ABCB1 3435C>T, and alpha-1 acid glycoprotein level, while (R)-methadone clearance was influenced by CYP2B6 activity, POR*28, and CYP3A4*22. Methadone 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-187 27125860-9 2016 The study also calls for caution related to higher exposure to 8-OH-EFV during simultaneous coadministration of EFV and RIF-based anti-TB regimens, which may be associated with neurotoxicity, particularly in female patients and CYP2B6*6 carriers. 8-oh-efv 63-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 228-234 27125860-9 2016 The study also calls for caution related to higher exposure to 8-OH-EFV during simultaneous coadministration of EFV and RIF-based anti-TB regimens, which may be associated with neurotoxicity, particularly in female patients and CYP2B6*6 carriers. efavirenz 68-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 228-234 27779789-4 2016 CNS toxicity occurring with the recommended standard dose of EFV remains a challenge and may in part be attributable to polymorphisms in cytochrome P450 (CYP) 2B6, the enzyme involved in the major metabolic pathway for converting EFV to inactive metabolites. efavirenz 61-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-162 27779789-5 2016 Functionally deficient alleles of CYP2B6 such as CYP2B6*6, *18, and *22 may be responsible for significantly higher therapeutic concentrations of EFV at a standard dose of 600 mg/day. efavirenz 146-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 27779789-5 2016 Functionally deficient alleles of CYP2B6 such as CYP2B6*6, *18, and *22 may be responsible for significantly higher therapeutic concentrations of EFV at a standard dose of 600 mg/day. efavirenz 146-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 27797496-4 2016 To improve our understanding of the molecular basis of the stimulatory effect of b5 and to test the hypothesis that b5 stimulates catalysis by more rapid protonation of the anionic ferric hydroperoxo heme intermediate of P450 (Fe3+OOH)- and subsequent formation of the active oxidizing species (Fe+4 O POR +), we have freeze-quenched the reaction mixture during a single turnover following reduction of oxyferrous P450 2B4 by each of its redox partners, b5 and P450 reductase. ferric hydroperoxo heme 181-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 221-225 27797496-4 2016 To improve our understanding of the molecular basis of the stimulatory effect of b5 and to test the hypothesis that b5 stimulates catalysis by more rapid protonation of the anionic ferric hydroperoxo heme intermediate of P450 (Fe3+OOH)- and subsequent formation of the active oxidizing species (Fe+4 O POR +), we have freeze-quenched the reaction mixture during a single turnover following reduction of oxyferrous P450 2B4 by each of its redox partners, b5 and P450 reductase. ferric sulfate 227-231 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 221-225 27797496-4 2016 To improve our understanding of the molecular basis of the stimulatory effect of b5 and to test the hypothesis that b5 stimulates catalysis by more rapid protonation of the anionic ferric hydroperoxo heme intermediate of P450 (Fe3+OOH)- and subsequent formation of the active oxidizing species (Fe+4 O POR +), we have freeze-quenched the reaction mixture during a single turnover following reduction of oxyferrous P450 2B4 by each of its redox partners, b5 and P450 reductase. OOH 231-234 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 221-225 27797496-4 2016 To improve our understanding of the molecular basis of the stimulatory effect of b5 and to test the hypothesis that b5 stimulates catalysis by more rapid protonation of the anionic ferric hydroperoxo heme intermediate of P450 (Fe3+OOH)- and subsequent formation of the active oxidizing species (Fe+4 O POR +), we have freeze-quenched the reaction mixture during a single turnover following reduction of oxyferrous P450 2B4 by each of its redox partners, b5 and P450 reductase. Iron 227-229 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 221-225 27797496-4 2016 To improve our understanding of the molecular basis of the stimulatory effect of b5 and to test the hypothesis that b5 stimulates catalysis by more rapid protonation of the anionic ferric hydroperoxo heme intermediate of P450 (Fe3+OOH)- and subsequent formation of the active oxidizing species (Fe+4 O POR +), we have freeze-quenched the reaction mixture during a single turnover following reduction of oxyferrous P450 2B4 by each of its redox partners, b5 and P450 reductase. 4 o por + 298-307 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 221-225 27749294-2 2016 Because genetic variations of propofol-metabolizing enzymes are proposed to be causal factors, we explored genetic polymorphisms of cytochrome P450 2B6 (CYP2B6) and uridine 5"-diphospho-glucuronosyltransferase 1A9 (UGT1A9). Propofol 30-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-151 27749294-2 2016 Because genetic variations of propofol-metabolizing enzymes are proposed to be causal factors, we explored genetic polymorphisms of cytochrome P450 2B6 (CYP2B6) and uridine 5"-diphospho-glucuronosyltransferase 1A9 (UGT1A9). Propofol 30-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 27749294-3 2016 Suggested high-risk factors (advanced age, CYP2B6 516 G/T, and UGT1A9 I399 C/C) were observed in this case of delayed propofol metabolism. Propofol 118-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 27917125-6 2016 After treatment with 20 muM rifampicin, mRNA increased 3- to 50-fold for all CYPs except CYP1A2 and 2D6 for HepaRG and 3D-PHH, 4-fold (CYP2B6) and 17-fold (CYP3A4) for 2D-PHH and four-fold (CYP3A4) for HepG2. Rifampin 28-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 27528039-7 2016 In contrast, pregnancy-induced increase in CYP2B6-catalyzed bupropion hydroxylation did not impact the plasma levels of OHBUP, probably due to a higher rate of OHBUP glucuronidation, and renal elimination associated with pregnancy. Bupropion 60-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 27865701-9 2016 The combination of the three CYP2B6 polymorphisms (G516T, C777A & A785G) revealed seven haplotypes. Adenosine Monophosphate 65-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 27528039-4 2016 Therefore, the aims of this study were to determine the steady-state pharmacokinetics of bupropion during pregnancy and the effect of functional genetic variants of CYP2B6 and CYP2C19 on bupropion pharmacokinetics in pregnant women. Bupropion 187-196 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-171 27538916-2 2016 To test this hypothesis, we investigated the effects of mutating these residues to Ala on the regiospecificity of CYP2B6 for the metabolism of testosterone and androstenedione. Alanine 83-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 27538916-2 2016 To test this hypothesis, we investigated the effects of mutating these residues to Ala on the regiospecificity of CYP2B6 for the metabolism of testosterone and androstenedione. Testosterone 143-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 27538916-2 2016 To test this hypothesis, we investigated the effects of mutating these residues to Ala on the regiospecificity of CYP2B6 for the metabolism of testosterone and androstenedione. Androstenedione 160-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 27538916-7 2016 Our results demonstrate that Phe206 plays a crucial role in determining the specificity of CYP2B6 for the 16beta-hydroxylation of testosterone and androstenedione and that it also plays an important role in BG binding and mechanism-based inactivation by BPA. Testosterone 130-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 27538916-7 2016 Our results demonstrate that Phe206 plays a crucial role in determining the specificity of CYP2B6 for the 16beta-hydroxylation of testosterone and androstenedione and that it also plays an important role in BG binding and mechanism-based inactivation by BPA. Androstenedione 147-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 27538916-7 2016 Our results demonstrate that Phe206 plays a crucial role in determining the specificity of CYP2B6 for the 16beta-hydroxylation of testosterone and androstenedione and that it also plays an important role in BG binding and mechanism-based inactivation by BPA. bisphenol A 254-257 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 27538916-8 2016 In addition, Val367 dramatically enhances the catalytic activity of CYP2B6 toward androstenedione and plays an important role in mechanism-based inactivation by BPA. Androstenedione 82-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 27538916-8 2016 In addition, Val367 dramatically enhances the catalytic activity of CYP2B6 toward androstenedione and plays an important role in mechanism-based inactivation by BPA. bisphenol A 161-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 27636207-7 2016 Liquid chromatography-time-of-flight mass spectrometry (LC-TOF/MS) of serum and urine revealed MDMA, 3,4-DMA, and the CYP-2B6 MDMA metabolites 3,4-methylendioxyamphetamine (MDA) and 4-hydroxy-3-methoxyamphetamine (HMA). N-Methyl-3,4-methylenedioxyamphetamine 126-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-125 27636207-7 2016 Liquid chromatography-time-of-flight mass spectrometry (LC-TOF/MS) of serum and urine revealed MDMA, 3,4-DMA, and the CYP-2B6 MDMA metabolites 3,4-methylendioxyamphetamine (MDA) and 4-hydroxy-3-methoxyamphetamine (HMA). 3,4-methylendioxyamphetamine 143-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-125 27636207-7 2016 Liquid chromatography-time-of-flight mass spectrometry (LC-TOF/MS) of serum and urine revealed MDMA, 3,4-DMA, and the CYP-2B6 MDMA metabolites 3,4-methylendioxyamphetamine (MDA) and 4-hydroxy-3-methoxyamphetamine (HMA). mda 173-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-125 27636207-7 2016 Liquid chromatography-time-of-flight mass spectrometry (LC-TOF/MS) of serum and urine revealed MDMA, 3,4-DMA, and the CYP-2B6 MDMA metabolites 3,4-methylendioxyamphetamine (MDA) and 4-hydroxy-3-methoxyamphetamine (HMA). 3-O-methyl-alpha-methyldopamine 182-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-125 27636207-7 2016 Liquid chromatography-time-of-flight mass spectrometry (LC-TOF/MS) of serum and urine revealed MDMA, 3,4-DMA, and the CYP-2B6 MDMA metabolites 3,4-methylendioxyamphetamine (MDA) and 4-hydroxy-3-methoxyamphetamine (HMA). 3-O-methyl-alpha-methyldopamine 214-217 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-125 27600044-0 2016 Inhibition of Cytochrome P450 2B6 Activity by Voriconazole Profiled Using Efavirenz Disposition in Healthy Volunteers. Voriconazole 46-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-33 27600044-3 2016 Efavirenz, a nonnucleoside HIV-1 reverse transcriptase inhibitor, is mainly cleared by CYP2B6, an enzyme strongly inhibited in vitro by voriconazole. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 27600044-3 2016 Efavirenz, a nonnucleoside HIV-1 reverse transcriptase inhibitor, is mainly cleared by CYP2B6, an enzyme strongly inhibited in vitro by voriconazole. Voriconazole 136-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 27600044-4 2016 To test efavirenz metabolism as an in vivo probe of CYP2B6 activity, we quantified the inhibition of CYP2B6 activity by voriconazole in 61 healthy volunteers administered a single 100-mg oral dose of efavirenz with and without voriconazole administration. Voriconazole 120-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 27600044-9 2016 This study demonstrates the mechanisms of voriconazole-efavirenz interaction, establishes the use of a low dose of efavirenz as a safe and selective in vivo probe for phenotyping CYP2B6 activity, and identifies several easy-to-use indices that should enhance understanding of the mechanisms of CYP2B6 interindividual variability. Voriconazole 42-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 179-185 27600044-9 2016 This study demonstrates the mechanisms of voriconazole-efavirenz interaction, establishes the use of a low dose of efavirenz as a safe and selective in vivo probe for phenotyping CYP2B6 activity, and identifies several easy-to-use indices that should enhance understanding of the mechanisms of CYP2B6 interindividual variability. Voriconazole 42-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 294-300 27457783-10 2016 NAC and selective inhibitors of CYP2B6 and CYP3A4 significantly reduced TMP covalent binding. Acetylcysteine 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 27477788-1 2016 Glycopeptide antibiotic biosynthesis involves a complex cascade of reactions centred on a non-ribosomal peptide synthetase and modifiying proteins acting in trans, such as Cytochrome P450 enzymes. Glycopeptides 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 27422672-6 2016 Although bupropion and efavirenz have been used and are recommended by regulatory agencies as clinical CYP2B6 probe substrates for DDI studies, CYP3A4 contributes to the metabolism of both probes and is induced by all reference CYP2B6 inducers. Bupropion 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 27422672-6 2016 Although bupropion and efavirenz have been used and are recommended by regulatory agencies as clinical CYP2B6 probe substrates for DDI studies, CYP3A4 contributes to the metabolism of both probes and is induced by all reference CYP2B6 inducers. efavirenz 23-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 27457783-10 2016 NAC and selective inhibitors of CYP2B6 and CYP3A4 significantly reduced TMP covalent binding. Trimethoprim 72-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 27495292-5 2016 In humans, the fraction of bupropion metabolized (fm) to the CYP2B6 probe metabolite OH-bupropion is 5-16%, but ticlopidine increases bupropion exposure by 61%, suggesting a 40% CYP2B6 and/or CYP2C19 fm for bupropion. Bupropion 27-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 27495292-5 2016 In humans, the fraction of bupropion metabolized (fm) to the CYP2B6 probe metabolite OH-bupropion is 5-16%, but ticlopidine increases bupropion exposure by 61%, suggesting a 40% CYP2B6 and/or CYP2C19 fm for bupropion. Bupropion 27-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-184 27457784-5 2016 However, negligible quantities of M11 and M12 were detected when EPA was incubated with a panel of human microsomes from multiple tissues, hepatocytes, recombinant human cytochrome P450s (P450s), and non-P450 enzymatic systems. epacadostat 65-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-185 27495292-5 2016 In humans, the fraction of bupropion metabolized (fm) to the CYP2B6 probe metabolite OH-bupropion is 5-16%, but ticlopidine increases bupropion exposure by 61%, suggesting a 40% CYP2B6 and/or CYP2C19 fm for bupropion. oh-bupropion 85-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 27495292-5 2016 In humans, the fraction of bupropion metabolized (fm) to the CYP2B6 probe metabolite OH-bupropion is 5-16%, but ticlopidine increases bupropion exposure by 61%, suggesting a 40% CYP2B6 and/or CYP2C19 fm for bupropion. Bupropion 88-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 27495292-5 2016 In humans, the fraction of bupropion metabolized (fm) to the CYP2B6 probe metabolite OH-bupropion is 5-16%, but ticlopidine increases bupropion exposure by 61%, suggesting a 40% CYP2B6 and/or CYP2C19 fm for bupropion. Bupropion 88-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 26141406-0 2016 Impact of the Cytochrome P450 2B6 (CYP2B6) Gene Polymorphism c.516G>T (rs3745274) on Propofol Dose Variability. Propofol 88-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-33 26141406-0 2016 Impact of the Cytochrome P450 2B6 (CYP2B6) Gene Polymorphism c.516G>T (rs3745274) on Propofol Dose Variability. Propofol 88-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 27439448-3 2016 CYP2B6 activity was measured by bupropion hydroxylation with LC/MS/MS. Bupropion 32-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26141406-2 2016 The present study sought to investigate the relationship between the c.516G>T polymorphism in the CYP2B6 (cytochrome P450 2B6) gene and the required propofol dose. Propofol 152-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 27439448-4 2016 The ratio of hydroxybupropion versus bupropion (AUC_hyd/AUC_bup) in terms of area under the time-concentration curve (AUC) was used to represent the CYP2B6 activity. hydroxybupropion 13-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 26141406-2 2016 The present study sought to investigate the relationship between the c.516G>T polymorphism in the CYP2B6 (cytochrome P450 2B6) gene and the required propofol dose. Propofol 152-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-128 27439448-4 2016 The ratio of hydroxybupropion versus bupropion (AUC_hyd/AUC_bup) in terms of area under the time-concentration curve (AUC) was used to represent the CYP2B6 activity. Bupropion 20-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 26141406-9 2016 CONCLUSIONS: Our results indicate that 34 % of the variance in the required propofol dose may be explained by these factors and that CYP2B6 c.516G>T polymorphism, which decreases the metabolism of the drug, accounts for approximately 7 % of the drug dosage. Propofol 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 26141406-10 2016 Our results show the possible influence of CYP2B6 c.516G>T genetic variant on propofol dose in patients under general anaesthesia. Propofol 81-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 27333947-10 2016 CONCLUSIONS: This study demonstrates that homozygous carriers of CYP2B6*6 allele may be at increased risk for EFV-induced QTcF interval prolongation via inhibition of hERG. efavirenz 110-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 27333947-2 2016 Since EFV is metabolized by the cytochrome P450 (CYP) 2B6 enzyme, we hypothesized that EFV would lengthen the rate-corrected QT (QTcF) interval in carriers of the CYP2B6*6 decreased functional allele. efavirenz 6-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-57 27627692-9 2016 On the other hand, HIV patients of African origin are predisposed to developing EFV-induced neuropsychiatric AEs due to specific CYP2B6 genetic variants causing impaired metabolism of EFV. efavirenz 80-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 27333947-2 2016 Since EFV is metabolized by the cytochrome P450 (CYP) 2B6 enzyme, we hypothesized that EFV would lengthen the rate-corrected QT (QTcF) interval in carriers of the CYP2B6*6 decreased functional allele. efavirenz 6-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 27333947-2 2016 Since EFV is metabolized by the cytochrome P450 (CYP) 2B6 enzyme, we hypothesized that EFV would lengthen the rate-corrected QT (QTcF) interval in carriers of the CYP2B6*6 decreased functional allele. efavirenz 87-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-57 27333947-2 2016 Since EFV is metabolized by the cytochrome P450 (CYP) 2B6 enzyme, we hypothesized that EFV would lengthen the rate-corrected QT (QTcF) interval in carriers of the CYP2B6*6 decreased functional allele. efavirenz 87-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 27333947-7 2016 RESULTS: EFV demonstrated a gene-dose effect and exceeded the FDA criteria for QTcF interval prolongation in CYP2B6*6/*6 carriers. efavirenz 9-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 27627692-9 2016 On the other hand, HIV patients of African origin are predisposed to developing EFV-induced neuropsychiatric AEs due to specific CYP2B6 genetic variants causing impaired metabolism of EFV. efavirenz 184-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 27281544-4 2016 In this work, a computational approach was carried out to study the interactions of 41 POPs (17 PBDEs, 17 PCBs, and 7 Dioxins) with four CYP2B6 protein structures downloaded from PDB data base (PDB: 3UA5, 3QOA, 3QU8 and 4I91) using molecular docking protocols with AutoDock Vina. Polychlorinated Biphenyls 106-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 27281544-2 2016 CYP2B6, a member of CYP450, is well known for being a highly inducible and polymorphic enzyme and for its important role in the oxidative metabolism of environmental pollutants, such as the Polybrominated Diphenyl Ethers (PBDEs) and Polychlorinated Biphenyls (PCBs). Halogenated Diphenyl Ethers 222-227 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27709010-5 2016 CYP2B6 has been demonstrated to play a role in the metabolism of 2%-10% of clinically used drugs including widely used antineoplastic agents cyclophosphamide and ifosfamide, anesthetics propofol and ketamine, synthetic opioids pethidine and methadone, and the antiretrovirals nevirapine and efavirenz, among others. Cyclophosphamide 141-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27709010-5 2016 CYP2B6 has been demonstrated to play a role in the metabolism of 2%-10% of clinically used drugs including widely used antineoplastic agents cyclophosphamide and ifosfamide, anesthetics propofol and ketamine, synthetic opioids pethidine and methadone, and the antiretrovirals nevirapine and efavirenz, among others. Ifosfamide 162-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27709010-5 2016 CYP2B6 has been demonstrated to play a role in the metabolism of 2%-10% of clinically used drugs including widely used antineoplastic agents cyclophosphamide and ifosfamide, anesthetics propofol and ketamine, synthetic opioids pethidine and methadone, and the antiretrovirals nevirapine and efavirenz, among others. Propofol 186-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27709010-5 2016 CYP2B6 has been demonstrated to play a role in the metabolism of 2%-10% of clinically used drugs including widely used antineoplastic agents cyclophosphamide and ifosfamide, anesthetics propofol and ketamine, synthetic opioids pethidine and methadone, and the antiretrovirals nevirapine and efavirenz, among others. Ketamine 199-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27709010-5 2016 CYP2B6 has been demonstrated to play a role in the metabolism of 2%-10% of clinically used drugs including widely used antineoplastic agents cyclophosphamide and ifosfamide, anesthetics propofol and ketamine, synthetic opioids pethidine and methadone, and the antiretrovirals nevirapine and efavirenz, among others. Meperidine 227-236 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27709010-5 2016 CYP2B6 has been demonstrated to play a role in the metabolism of 2%-10% of clinically used drugs including widely used antineoplastic agents cyclophosphamide and ifosfamide, anesthetics propofol and ketamine, synthetic opioids pethidine and methadone, and the antiretrovirals nevirapine and efavirenz, among others. Methadone 241-250 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27709010-5 2016 CYP2B6 has been demonstrated to play a role in the metabolism of 2%-10% of clinically used drugs including widely used antineoplastic agents cyclophosphamide and ifosfamide, anesthetics propofol and ketamine, synthetic opioids pethidine and methadone, and the antiretrovirals nevirapine and efavirenz, among others. Nevirapine 276-286 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27709010-5 2016 CYP2B6 has been demonstrated to play a role in the metabolism of 2%-10% of clinically used drugs including widely used antineoplastic agents cyclophosphamide and ifosfamide, anesthetics propofol and ketamine, synthetic opioids pethidine and methadone, and the antiretrovirals nevirapine and efavirenz, among others. efavirenz 291-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27281544-2 2016 CYP2B6, a member of CYP450, is well known for being a highly inducible and polymorphic enzyme and for its important role in the oxidative metabolism of environmental pollutants, such as the Polybrominated Diphenyl Ethers (PBDEs) and Polychlorinated Biphenyls (PCBs). Polychlorinated Biphenyls 233-258 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27281544-2 2016 CYP2B6, a member of CYP450, is well known for being a highly inducible and polymorphic enzyme and for its important role in the oxidative metabolism of environmental pollutants, such as the Polybrominated Diphenyl Ethers (PBDEs) and Polychlorinated Biphenyls (PCBs). Polychlorinated Biphenyls 260-264 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27281544-3 2016 However the mechanisms of interaction of PBDEs and PCBs with CYP2B6 is not entirely known. Halogenated Diphenyl Ethers 41-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 27281544-3 2016 However the mechanisms of interaction of PBDEs and PCBs with CYP2B6 is not entirely known. Polychlorinated Biphenyls 51-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 27281544-4 2016 In this work, a computational approach was carried out to study the interactions of 41 POPs (17 PBDEs, 17 PCBs, and 7 Dioxins) with four CYP2B6 protein structures downloaded from PDB data base (PDB: 3UA5, 3QOA, 3QU8 and 4I91) using molecular docking protocols with AutoDock Vina. Halogenated Diphenyl Ethers 96-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 27281544-0 2016 Mechanisms of interaction between persistent organic pollutants (POPs) and CYP2B6: An in silico approach. POPS 65-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 27281544-4 2016 In this work, a computational approach was carried out to study the interactions of 41 POPs (17 PBDEs, 17 PCBs, and 7 Dioxins) with four CYP2B6 protein structures downloaded from PDB data base (PDB: 3UA5, 3QOA, 3QU8 and 4I91) using molecular docking protocols with AutoDock Vina. Dioxins 118-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 27281544-5 2016 The best binding affinity values (kcal/mol) were obtained for PBDE-99 (-8.5), PCB-187 (-9.6), and octachloro-dibenzo-dioxin (-9.8) that can be attributed to the hydrophobic interactions with important residues, such as Phe-363, in the catalytic site of CYP2B6. 2,2',4,4',5-brominated diphenyl ether 62-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 253-259 27281544-2 2016 CYP2B6, a member of CYP450, is well known for being a highly inducible and polymorphic enzyme and for its important role in the oxidative metabolism of environmental pollutants, such as the Polybrominated Diphenyl Ethers (PBDEs) and Polychlorinated Biphenyls (PCBs). Phenyl Ethers 205-220 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27281544-5 2016 The best binding affinity values (kcal/mol) were obtained for PBDE-99 (-8.5), PCB-187 (-9.6), and octachloro-dibenzo-dioxin (-9.8) that can be attributed to the hydrophobic interactions with important residues, such as Phe-363, in the catalytic site of CYP2B6. 2,2',3,4',5,5',6-HEPTACHLOROBIPHENYL 78-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 253-259 27867424-0 2016 Aldehyde and Ketone Synthesis by P450-Catalyzed Oxidative Deamination of Alkyl Azides. Aldehydes 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 27281544-5 2016 The best binding affinity values (kcal/mol) were obtained for PBDE-99 (-8.5), PCB-187 (-9.6), and octachloro-dibenzo-dioxin (-9.8) that can be attributed to the hydrophobic interactions with important residues, such as Phe-363, in the catalytic site of CYP2B6. octachlorodibenzo-4-dioxin 98-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 253-259 27281544-5 2016 The best binding affinity values (kcal/mol) were obtained for PBDE-99 (-8.5), PCB-187 (-9.6), and octachloro-dibenzo-dioxin (-9.8) that can be attributed to the hydrophobic interactions with important residues, such as Phe-363, in the catalytic site of CYP2B6. Phenylalanine 219-222 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 253-259 27904432-0 2016 Corrections of Frequencies of Cytochrome P450 2B6 and 2C8 Allelic Variants in the Mozambican Population. mozambican 82-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-57 26711482-0 2016 Evaluation of cytochrome P450 inductions by anti-epileptic drug oxcarbazepine, 10-hydroxyoxcarbazepine, and carbamazepine using human hepatocytes and HepaRG cells. Oxcarbazepine 64-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 26711482-0 2016 Evaluation of cytochrome P450 inductions by anti-epileptic drug oxcarbazepine, 10-hydroxyoxcarbazepine, and carbamazepine using human hepatocytes and HepaRG cells. 10-hydroxyoxcarbazepine 79-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 26711482-0 2016 Evaluation of cytochrome P450 inductions by anti-epileptic drug oxcarbazepine, 10-hydroxyoxcarbazepine, and carbamazepine using human hepatocytes and HepaRG cells. Carbamazepine 108-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 26711482-2 2016 Auto-induction potentials of oxcarbazepine, its pharmacologically active metabolite 10-hydroxyoxcarbazepine and carbamazepine were evaluated by cytochrome P450 (CYP) 1A2, CYP2B6 and CYP3A4 mRNA levels and primary metabolic rates using human hepatocytes and HepaRG cells. Oxcarbazepine 29-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 26711482-4 2016 The fold change in mRNA levels for CYP2B6 was 11.5 (3.2-19.3), 7.0 (2.5-10.8) and 14.8 (3.1-29.1) for oxcarbazepine, 10-hydroxyoxcarbazepine and carbamazepine, respectively. Oxcarbazepine 102-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 26711482-4 2016 The fold change in mRNA levels for CYP2B6 was 11.5 (3.2-19.3), 7.0 (2.5-10.8) and 14.8 (3.1-29.1) for oxcarbazepine, 10-hydroxyoxcarbazepine and carbamazepine, respectively. 10-hydroxyoxcarbazepine 117-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 26711482-4 2016 The fold change in mRNA levels for CYP2B6 was 11.5 (3.2-19.3), 7.0 (2.5-10.8) and 14.8 (3.1-29.1) for oxcarbazepine, 10-hydroxyoxcarbazepine and carbamazepine, respectively. Carbamazepine 145-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 26711482-9 2016 The 10-keto group instead of double bond at C10 position is evidently a determinant factor for limited auto-induction of P450 enzymes by oxcarbazepine. Oxcarbazepine 137-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 27028535-4 2016 It undergoes oxidative metabolism, mainly to norketamine by cytochrome P450 (CYP) 3A and CYP2B6 enzymes. norketamine 45-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-95 27402728-2 2016 An efficient and stereospecific enzymatic synthesis of 1beta-OH-DCA was developed using a Bacillus megaterium (BM3) cytochrome P450 (P450) mutant, and its structure was confirmed by nuclear magnetic resonance (NMR) spectroscopy. 1beta-oh-dca 55-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-137 27402728-6 2016 Screening of 21 recombinant human cytochrome P450 (P450) enzymes showed that, with the exception of extrahepatic CYP46A1, the most abundant liver P450 subfamily CYP3A, including CYP3A4, 3A5, and 3A7, specifically catalyzed 1beta-OH-DCA formation. 1beta-oh-dca 223-235 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-55 27867424-0 2016 Aldehyde and Ketone Synthesis by P450-Catalyzed Oxidative Deamination of Alkyl Azides. Ketones 13-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 27867424-0 2016 Aldehyde and Ketone Synthesis by P450-Catalyzed Oxidative Deamination of Alkyl Azides. alkyl azides 73-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 27867424-3 2016 Our studies show that although several heme-containing enzymes possess basal activity in this reaction, an engineered variant of the bacterial cytochrome P450 CYP102A1 constitutes a particularly efficient biocatalyst for promoting this transformation, exhibiting a broad substrate scope along with high catalytic activity (up to 11,300 TON), excellent chemoselectivity, and enhanced reactivity toward secondary alkyl azides to yield ketones. Heme 39-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 27867424-3 2016 Our studies show that although several heme-containing enzymes possess basal activity in this reaction, an engineered variant of the bacterial cytochrome P450 CYP102A1 constitutes a particularly efficient biocatalyst for promoting this transformation, exhibiting a broad substrate scope along with high catalytic activity (up to 11,300 TON), excellent chemoselectivity, and enhanced reactivity toward secondary alkyl azides to yield ketones. alkyl azides 411-423 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 27867424-3 2016 Our studies show that although several heme-containing enzymes possess basal activity in this reaction, an engineered variant of the bacterial cytochrome P450 CYP102A1 constitutes a particularly efficient biocatalyst for promoting this transformation, exhibiting a broad substrate scope along with high catalytic activity (up to 11,300 TON), excellent chemoselectivity, and enhanced reactivity toward secondary alkyl azides to yield ketones. Ketones 433-440 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 27867424-5 2016 Altogether, these studies demonstrate that engineered P450 variants represent promising biocatalysts for the synthesis of aryl aldehydes and ketones via the oxidative deamination of alkyl azides under mild reaction conditions. aryl aldehydes 122-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 27867424-5 2016 Altogether, these studies demonstrate that engineered P450 variants represent promising biocatalysts for the synthesis of aryl aldehydes and ketones via the oxidative deamination of alkyl azides under mild reaction conditions. Ketones 141-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 27867424-5 2016 Altogether, these studies demonstrate that engineered P450 variants represent promising biocatalysts for the synthesis of aryl aldehydes and ketones via the oxidative deamination of alkyl azides under mild reaction conditions. alkyl azides 182-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 27339894-1 2016 Cytochrome P450 (P450) reactions can involve C-C bond cleavage, and several of these are critical in steroid and sterol biosynthesis. Steroids 101-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 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 2 subfamily B member 6 Homo sapiens 27-33 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 2 subfamily B member 6 Homo sapiens 282-288 27339894-1 2016 Cytochrome P450 (P450) reactions can involve C-C bond cleavage, and several of these are critical in steroid and sterol biosynthesis. Sterols 113-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 27278961-4 2016 The exception was C-7, a hydroxylated metabolite, largely formed by CYP2B6 and CYP3A4/5. Carbon 18-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 27441453-6 2016 Using menadione as the redox-cycling chemical, we discovered that this enzymatic reaction blocks metabolic activation of parathion in rat and human liver microsomes and in recombinant CYPs important to parathion metabolism, including CYP1A2, CYP2B6, and CYP3A4. Vitamin K 3 6-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-248 27441453-6 2016 Using menadione as the redox-cycling chemical, we discovered that this enzymatic reaction blocks metabolic activation of parathion in rat and human liver microsomes and in recombinant CYPs important to parathion metabolism, including CYP1A2, CYP2B6, and CYP3A4. Parathion 121-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-248 27208383-0 2016 Establishment of In Silico Prediction Models for CYP3A4 and CYP2B6 Induction in Human Hepatocytes by Multiple Regression Analysis Using Azole Compounds. Azoles 136-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 27208383-3 2016 In this study, we have established regression models for CYP3A4 and CYP2B6 induction in human hepatocytes using several physicochemical parameters for a set of azole compounds with different P450 induction as characteristics as model compounds. Azoles 160-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 27208383-7 2016 The fold-induction of the validation compounds, another set of 12 azole-containing compounds, were predicted within twofold limits for both CYP3A4 and CYP2B6. Azoles 66-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 26626330-10 2016 Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Nevirapine 124-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 26626330-10 2016 Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Ritonavir 146-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 27311985-8 2016 Transfection of HepaRG cells with hsa-miR-25-3p mimics inhibited expression of the endogenous CYP2B6 gene and it also decreased rifampicin-dependent induction of CYP2B6 at the mRNA and protein levels. Rifampin 128-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 27197997-6 2016 Activation of CAR and PXR in cells treated with a high dose of CITCO [6-(4-chlorophenyl)-imidazo(2,1-b)thiazole-5-carbaldehyde] or cotreated with rifampicin and tetracycline resulted in synergistic enhancement of CYP3A4, but not CYP2B6, CYP2C9, or UGT1A1, mRNA expression in HepTR/hCAR/hPXR cells. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 63-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 229-235 27197997-6 2016 Activation of CAR and PXR in cells treated with a high dose of CITCO [6-(4-chlorophenyl)-imidazo(2,1-b)thiazole-5-carbaldehyde] or cotreated with rifampicin and tetracycline resulted in synergistic enhancement of CYP3A4, but not CYP2B6, CYP2C9, or UGT1A1, mRNA expression in HepTR/hCAR/hPXR cells. 6-(4-chlorophenyl)-imidazo(2,1-b)thiazole 70-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 229-235 27289271-0 2016 Relationship between CYP2B6*6 and cold pressor pain sensitivity in opioid dependent patients on methadone maintenance therapy (MMT). Methadone 96-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). cb118 38-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-83 27289271-0 2016 Relationship between CYP2B6*6 and cold pressor pain sensitivity in opioid dependent patients on methadone maintenance therapy (MMT). mmt 127-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 27289271-1 2016 BACKGROUND: CYP2B6 polymorphisms contribute to inter-individual variations in pharmacokinetics of methadone. Methadone 98-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 27289271-3 2016 It is possible, therefore, that genetic polymorphisms in CYP2B6, which affects the metabolism of methadone, influence pain sensitivity among patients on MMT. Methadone 97-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 27289271-3 2016 It is possible, therefore, that genetic polymorphisms in CYP2B6, which affects the metabolism of methadone, influence pain sensitivity among patients on MMT. mmt 153-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 27289271-10 2016 CONCLUSION: Our study indicates that the CYP2B6*6 allele is associated with a lower pain threshold and lower pain tolerance among males with opioid dependence on MMT. mmt 162-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 27289271-11 2016 The CYP2B6*6 allele may provide a mechanistic explanation for clinical observations of heightened pain sensitivity among opioid dependent patients receiving MMT. mmt 157-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). 4-oh-cb107 260-270 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-83 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). 4-oh-cb107 260-270 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). cb118 38-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). 3-hydroxy (oh)-cb118 155-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-83 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). 3-hydroxy (oh)-cb118 155-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). cb118 170-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-83 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). cb118 170-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl 217-258 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-83 27208082-4 2016 In vitro studies on the metabolism of CB118 by cytochrome P450 enzymes (P450 or CYP) revealed that human CYP2B6 and rat CYP2B1 primarily metabolized it to 3-hydroxy (OH)-CB118, whereas rat CYP1A1 metabolized CB118 to 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl (4-OH-CB107). 4-hydroxy-2,3,3",4",5-pentachlorobiphenyl 217-258 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 27296047-8 2016 The total WEA extract inhibited CYP2B6 (OH-BUP) and CYP2C19 (5-OH-OME) with high affinity, IC50 ~ 20 mug ml(-1), while moderate inhibitory interactions were observed for CYP1A2, CYP2D6, CYP2E1 and CYP3A4. oh-bup 40-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 27434302-5 2016 METHODS: To investigate this we used a drug cocktail validated in humans consisting of five widely prescribed drugs as probes for specific P450 enzymes: caffeine (CYP1A2), metoprolol (CYP2D6), omeprazole (CYP2C19), midazolam (CYP3A4) and s-warfarin (CYP2C9). Caffeine 153-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 139-143 27434302-5 2016 METHODS: To investigate this we used a drug cocktail validated in humans consisting of five widely prescribed drugs as probes for specific P450 enzymes: caffeine (CYP1A2), metoprolol (CYP2D6), omeprazole (CYP2C19), midazolam (CYP3A4) and s-warfarin (CYP2C9). Midazolam 215-224 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 139-143 27434302-5 2016 METHODS: To investigate this we used a drug cocktail validated in humans consisting of five widely prescribed drugs as probes for specific P450 enzymes: caffeine (CYP1A2), metoprolol (CYP2D6), omeprazole (CYP2C19), midazolam (CYP3A4) and s-warfarin (CYP2C9). Warfarin 238-248 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 139-143 27434302-14 2016 The changes in drug clearance correlated with the expression pattern of the specific P450 enzymes in case of Cyp1a2-caffeine and Cyp2c37-omeprazole. Caffeine 116-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 27434302-14 2016 The changes in drug clearance correlated with the expression pattern of the specific P450 enzymes in case of Cyp1a2-caffeine and Cyp2c37-omeprazole. Omeprazole 137-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 27195527-0 2016 Pregnancy affects nevirapine pharmacokinetics: evidence from a CYP2B6 genotype-guided observational study. Nevirapine 18-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 27195527-3 2016 RESULTS: CYP2B6 516 G>T and 983 T>C were associated independently with plasma nevirapine concentrations in pregnant (n=110) and postpartum (n=122) women and were used for stratification. Nevirapine 84-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 27388155-0 2016 CYP2B6rs2279343 Is Associated with Improved Survival of Pediatric Rhabdomyosarcoma Treated with Cyclophosphamide. Cyclophosphamide 96-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27388155-3 2016 CYP2B6 is a highly polymorphic drug metabolizing enzyme involved in CPA bioactivation. Cyclophosphamide 68-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27388155-5 2016 METHODS: We genotyped CYP2B6SNPs rs2279343, rs3745274, and rs3211371 by restriction fragment polymorphism (RFLP) after PCR amplification in a cohort of 73 pediatric RMS patients treated with CPA-based first line treatment. Cyclophosphamide 191-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 27388155-10 2016 CONCLUSION: Our results demonstrated that CYP2B6 rs2279343 may predict EFS in RMS patients and warrants future studies to clarify the pharmacogenetics of CPA in pediatrics. Cyclophosphamide 154-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 27435752-6 2016 Next, independent DDI studies with substrates of CYP3A4 (maraviroc, atazanavir, and clarithromycin) and CYP2B6 (bupropion) verified the induction components of the model (area under the curve [AUC] ratios within 1.0-1.7-fold of observed). Bupropion 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-110 27185541-4 2016 The observed phase-I metabolites were formed through N-deethylation, N,N-deethylation, N-hydroxylation, and de-esterification, with CYP2B6 and CYP2C19 being the main enzymes catalyzing their formation. Nitrogen 53-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-138 27311637-5 2016 Moreover, berberine inhibits expressions of CAR and its target genes CYP2B6 and CYP3A4. Berberine 10-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 27052878-8 2016 In HLMs with lower CYP2D6 activity levels, 2-hydroxymethylatomoxetine (2-CH2OH-ATX) formation became a more predominant pathway of metabolism, which appeared to be catalyzed by CYP2B6. UNII-98EAX636XE 43-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-183 27052878-8 2016 In HLMs with lower CYP2D6 activity levels, 2-hydroxymethylatomoxetine (2-CH2OH-ATX) formation became a more predominant pathway of metabolism, which appeared to be catalyzed by CYP2B6. 2-ch2oh-atx 71-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-183 27362518-8 2016 CYP2B6 catalyzes propofol and ketamine metabolism. Propofol 17-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27362518-8 2016 CYP2B6 catalyzes propofol and ketamine metabolism. Ketamine 30-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27362518-9 2016 CYP2B6*6 allele is associated with decreased propofol and ketamine metabolism and increased adverse effects. Propofol 45-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27362518-9 2016 CYP2B6*6 allele is associated with decreased propofol and ketamine metabolism and increased adverse effects. Ketamine 58-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27660681-1 2016 Bupropion is a widely used antidepressant and the recommended CYP2B6 probe drug. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 26885802-10 2016 CYP2B6 poor metabolizers displayed higher EFV AUCtau and Cmax (125% and 91%, respectively) versus non-poor metabolizers, and lower EVG and COBI exposures. elvitegravir 131-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27329697-5 2016 In humans, plumbagin was not only a mixed inhibitor of CYP2B6, CYP2C9, CYP2D6, CYP2E1 and CYP3A4, but also a non-competitive inhibitor of CYP1A2, with Ki values no more than 2.16 muM. plumbagin 11-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 27311637-6 2016 Furthermore, berberine enhances DNA methylation level in whole genome but reduces that in promoter regions CpG sites of CYP2B6 and CYP3A4 genes under the presence of CAR condition. Berberine 13-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 27059013-4 2016 Purified recombinant hP450 27C1 bound and desaturated all-trans retinol, retinal, and retinoic acid, as well as 11-cis-retinal. Vitamin A 64-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-26 26940377-7 2016 Importantly, chalcone isomerase, isoflavone synthase, cytochrome p450, UDP-glycosyltransferase, and isoflavone reductase, which are all involved in the biosynthesis pathway of bioactive isoflavonoids, were most abundantly expressed in the summer-collected tubers. isoflavonoids 186-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 27067333-3 2016 The objective of this analysis was to support the selection of an appropriate dose for this younger pediatric population and to explore the impact of CYP2B6 genetic polymorphisms on EFV systemic exposures. efavirenz 182-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 26655325-9 2016 Compared to noncarriers, individuals homozygous for CYP2B6*6 had ~109% increased EFV levels in hair (p = .016) and CYP2B6*18 heterozygotes demonstrated 82% higher EFV hair levels (p = .0006). efavirenz 81-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 26655325-9 2016 Compared to noncarriers, individuals homozygous for CYP2B6*6 had ~109% increased EFV levels in hair (p = .016) and CYP2B6*18 heterozygotes demonstrated 82% higher EFV hair levels (p = .0006). efavirenz 163-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 26655325-10 2016 This study confirmed that alleles affecting CYP2B6 metabolism and subsequent EFV exposure are present at significant frequencies in both the SAB and CMA populations. sab 141-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 27059013-4 2016 Purified recombinant hP450 27C1 bound and desaturated all-trans retinol, retinal, and retinoic acid, as well as 11-cis-retinal. Tretinoin 86-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-26 27059013-5 2016 Although the physiological role of 3,4-dehydroretinoids in humans is unclear, we have identified hP450 27C1 as an enzyme capable of efficiently mediating their formation. 3,4-dehydroretinoids 35-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-102 27128896-4 2016 Aschantin at 100 microM negligibly inhibited CYP1A2-mediated phenacetin O-de-ethylation, CYP2A6-mediated coumarin 7-hydroxylation, CYP2B6-mediated bupropion hydroxylation, and CYP2D6-mediated bufuralol 1"-hydroxylation. Bupropion 147-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 26918316-1 2016 Cytochrome P450 (P450) enzymes metabolize arachidonic acid (AA) to several biologically active epoxyeicosatrienoic acids (EETs) and hydroxyeicosatetraenoic acids (HETEs). Arachidonic Acid 42-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 27128896-4 2016 Aschantin at 100 microM negligibly inhibited CYP1A2-mediated phenacetin O-de-ethylation, CYP2A6-mediated coumarin 7-hydroxylation, CYP2B6-mediated bupropion hydroxylation, and CYP2D6-mediated bufuralol 1"-hydroxylation. aschantin 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 26982502-6 2016 A structure of CYP2B6 Y244W in complex with (+)-alpha-pinene was solved at 2.2 A and showed no CYMAL-5 in the peripheral pocket. alpha-pinene 44-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 26918316-1 2016 Cytochrome P450 (P450) enzymes metabolize arachidonic acid (AA) to several biologically active epoxyeicosatrienoic acids (EETs) and hydroxyeicosatetraenoic acids (HETEs). epoxyeicosatrienoic acids 95-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 26918316-1 2016 Cytochrome P450 (P450) enzymes metabolize arachidonic acid (AA) to several biologically active epoxyeicosatrienoic acids (EETs) and hydroxyeicosatetraenoic acids (HETEs). eets 122-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 26918316-1 2016 Cytochrome P450 (P450) enzymes metabolize arachidonic acid (AA) to several biologically active epoxyeicosatrienoic acids (EETs) and hydroxyeicosatetraenoic acids (HETEs). Hydroxyeicosatetraenoic Acids 132-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 26918316-1 2016 Cytochrome P450 (P450) enzymes metabolize arachidonic acid (AA) to several biologically active epoxyeicosatrienoic acids (EETs) and hydroxyeicosatetraenoic acids (HETEs). Hydroxyeicosatetraenoic Acids 163-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 26369775-10 2016 Vonoprazan is metabolized to inactive metabolites mainly by cytochrome P450 (CYP)3A4 and to some extent by CYP2B6, CYP2C19, CYP2D6, and SULT2A1. 1-(5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 27045425-0 2016 CYP2B6 genotype-based efavirenz dose recommendations during rifampicin-based antituberculosis cotreatment for a sub-Saharan Africa population. Rifampin 60-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27010727-0 2016 Genome-Wide Pharmacogenomic Study on Methadone Maintenance Treatment Identifies SNP rs17180299 and Multiple Haplotypes on CYP2B6, SPON1, and GSG1L Associated with Plasma Concentrations of Methadone R- and S-enantiomers in Heroin-Dependent Patients. Methadone 37-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 27010727-0 2016 Genome-Wide Pharmacogenomic Study on Methadone Maintenance Treatment Identifies SNP rs17180299 and Multiple Haplotypes on CYP2B6, SPON1, and GSG1L Associated with Plasma Concentrations of Methadone R- and S-enantiomers in Heroin-Dependent Patients. Methadone 188-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 27010727-0 2016 Genome-Wide Pharmacogenomic Study on Methadone Maintenance Treatment Identifies SNP rs17180299 and Multiple Haplotypes on CYP2B6, SPON1, and GSG1L Associated with Plasma Concentrations of Methadone R- and S-enantiomers in Heroin-Dependent Patients. Heroin 222-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 27010727-10 2016 The association between the S-methadone plasma concentration and CYP2B6, SPON1, and GSG1L were replicated in another independent study. Methadone 28-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 26811874-6 2016 We report here the identification of a P450 [P450cam (CYP101A1) and P450cin (CYP176A1)]-generated arene oxide as a product of in vitro oxidation of tert-butylbenzene. Benzene oxide 98-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-52 26811874-6 2016 We report here the identification of a P450 [P450cam (CYP101A1) and P450cin (CYP176A1)]-generated arene oxide as a product of in vitro oxidation of tert-butylbenzene. tert-butylbenzene 148-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-52 26802129-1 2016 Bupropion is a widely used antidepressant and smoking cessation aid in addition to being one of two US Food and Drug Administration-recommended probe substrates for evaluation of cytochrome P450 2B6 activity. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 179-198 26830411-0 2016 Influence of CYP2B6 and CYP2C19 polymorphisms on sertraline metabolism in major depression patients. Sertraline 49-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 26830411-1 2016 BACKGROUND: Genetic polymorphisms in CYP2B6 and CYP2C19 may cause variability in the metabolism of sertraline, a widely used antidepressant in major depressive disorder treatment. Sertraline 99-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 26830411-11 2016 CONCLUSION: CYP2B6*6 and *9 variant alleles had a significant decreasing effect on sertraline metabolism in major depression patients which might result as variations in sertraline therapy. Sertraline 83-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 26830411-11 2016 CONCLUSION: CYP2B6*6 and *9 variant alleles had a significant decreasing effect on sertraline metabolism in major depression patients which might result as variations in sertraline therapy. Sertraline 170-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 27045425-4 2016 RESULTS: Simulated AUCs for 600 mg EFV dose were 1.2- and 2.4-times greater than the product label for Ugandans in general and CYP2B6*6/*6 genotypes respectively. efavirenz 35-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 27045425-5 2016 EFV daily doses of 450 and 250 mg for Ugandans and CYP2B6*6/*6 genotypes, respectively, yielded simulated exposures comparable to the product label. efavirenz 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 26823489-1 2016 The constitutive androstane receptor (CAR and NR1i3) is a key regulator of CYP2B6, the enzyme predominantly responsible for the biotransformation of cyclophosphamide (CPA) to its pharmacologically active metabolite, 4-hydroxycyclophosphamide (4-OH-CPA). Cyclophosphamide 149-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 26823489-1 2016 The constitutive androstane receptor (CAR and NR1i3) is a key regulator of CYP2B6, the enzyme predominantly responsible for the biotransformation of cyclophosphamide (CPA) to its pharmacologically active metabolite, 4-hydroxycyclophosphamide (4-OH-CPA). Cyclophosphamide 167-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 26823489-1 2016 The constitutive androstane receptor (CAR and NR1i3) is a key regulator of CYP2B6, the enzyme predominantly responsible for the biotransformation of cyclophosphamide (CPA) to its pharmacologically active metabolite, 4-hydroxycyclophosphamide (4-OH-CPA). 4-hydroxycyclophosphamide 216-241 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 26823489-1 2016 The constitutive androstane receptor (CAR and NR1i3) is a key regulator of CYP2B6, the enzyme predominantly responsible for the biotransformation of cyclophosphamide (CPA) to its pharmacologically active metabolite, 4-hydroxycyclophosphamide (4-OH-CPA). 4-oh-cpa 243-251 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 26456622-5 2016 RESULTS: Multiple linear regression analysis revealed that CYP2B6 -750 T > C (P < 0.001), -2320 T > C (P < 0.001), 15582C > T (P = 0.017), CYP2C19*2 (P < 0.001) and PXR 66034 T > C (P = 0.028) accounted for 47% of the variation in 4-OH-CPA plasma concentration. 4-oh-cpa 252-260 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 26891286-5 2016 Efavirenz (EFV) and nevirapine (NVP) are the major components of HAART both metabolized by CYP2B6, an enzyme that can potentially be inhibited or induced by compounds found in medicinal plant extracts. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 26891286-5 2016 Efavirenz (EFV) and nevirapine (NVP) are the major components of HAART both metabolized by CYP2B6, an enzyme that can potentially be inhibited or induced by compounds found in medicinal plant extracts. efavirenz 11-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 26891286-5 2016 Efavirenz (EFV) and nevirapine (NVP) are the major components of HAART both metabolized by CYP2B6, an enzyme that can potentially be inhibited or induced by compounds found in medicinal plant extracts. Nevirapine 20-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 26831894-2 2016 We describe four children presenting with serious, persistent central nervous system adverse events who were found to have elevated plasma efavirenz concentrations as a result of carrying CYP2B6 single nucleotide polymorphisms, known to play a role in the metabolism of EFV. efavirenz 270-273 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 188-194 26456622-6 2016 Among these variants, CYP2B6 -750 T > C and CYP2C19*2 were selected as the combination genetic marker because these two SNPs contributed the most to the inter-individual variability in 4-OH-CPA concentration, accounting for 23.6% and 21.5% of the variation, respectively. 4-oh-cpa 188-196 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 26456622-0 2016 Genetic markers in CYP2C19 and CYP2B6 for prediction of cyclophosphamide"s 4-hydroxylation, efficacy and side effects in Chinese patients with systemic lupus erythematosus. Cyclophosphamide 56-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 26763401-1 2016 The effects of three kinds of penicillin-based antibiotics, amoxicillin, ampicillin, and piperacillin, on drug-metabolizing activity of human hepatic cytochrome P450 (P450 or CYP) were investigated. Penicillins 30-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-178 26688241-6 2016 Moreover, the evaluation on the changes of metabolic enzymes revealed that HBCD and alpha-HCH shared a similar pattern of cytochrome P450 induction (CYP2B6), which was different from those of PCBs and BDE47 (CYP1A1 and CYP2B6). alpha-hexachlorocyclohexane 84-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 26688241-6 2016 Moreover, the evaluation on the changes of metabolic enzymes revealed that HBCD and alpha-HCH shared a similar pattern of cytochrome P450 induction (CYP2B6), which was different from those of PCBs and BDE47 (CYP1A1 and CYP2B6). alpha-hexachlorocyclohexane 84-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 219-225 25651810-6 2016 In vitro studies demonstrate that C-PCBs with a 2,3,6-trichlorosubstitution pattern in one phenyl ring are readily oxidized to hydroxylated PCB metabolites (HO-PCBs) by P450 enzymes, such as rat CYP2B1, human CYP2B6, and dog CYP2B11. c-pcbs 34-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 209-215 25651810-6 2016 In vitro studies demonstrate that C-PCBs with a 2,3,6-trichlorosubstitution pattern in one phenyl ring are readily oxidized to hydroxylated PCB metabolites (HO-PCBs) by P450 enzymes, such as rat CYP2B1, human CYP2B6, and dog CYP2B11. Polychlorinated Biphenyls 36-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 209-215 25651810-6 2016 In vitro studies demonstrate that C-PCBs with a 2,3,6-trichlorosubstitution pattern in one phenyl ring are readily oxidized to hydroxylated PCB metabolites (HO-PCBs) by P450 enzymes, such as rat CYP2B1, human CYP2B6, and dog CYP2B11. ho-pcbs 157-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 209-215 26201057-6 2016 Induction of CYP1A2, CYP2B6 and CYP3A4 activities in the tethered spheroids were comparable to, if not higher than that observed in the collagen sandwich cultures. spheroids 66-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 26763401-1 2016 The effects of three kinds of penicillin-based antibiotics, amoxicillin, ampicillin, and piperacillin, on drug-metabolizing activity of human hepatic cytochrome P450 (P450 or CYP) were investigated. Amoxicillin 60-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-178 26763401-1 2016 The effects of three kinds of penicillin-based antibiotics, amoxicillin, ampicillin, and piperacillin, on drug-metabolizing activity of human hepatic cytochrome P450 (P450 or CYP) were investigated. Ampicillin 73-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-178 26763401-1 2016 The effects of three kinds of penicillin-based antibiotics, amoxicillin, ampicillin, and piperacillin, on drug-metabolizing activity of human hepatic cytochrome P450 (P450 or CYP) were investigated. Piperacillin 89-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-178 26553012-1 2016 Efavirenz (EFV) is principally metabolized by CYP2B6 to 8-hydroxy-efavirenz (8OH-EFV) and to a lesser extent by CYP2A6 to 7-hydroxy-efavirenz (7OH-EFV). efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 25963334-0 2016 CYP2B6*6 genotype and high efavirenz plasma concentration but not nevirapine are associated with low lumefantrine plasma exposure and poor treatment response in HIV-malaria-coinfected patients. Lumefantrine 101-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 25963334-5 2016 High EFV plasma concentrations and CYP2B6*6/*6 genotype significantly correlated with low lumefantrine plasma concentrations and high rate of recurrent parasitemia. Lumefantrine 90-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 26779253-4 2015 The drug metabolizing enzyme (DME), CYP2B6, is primarily responsible for EFV metabolism with minor contributions by CYP1A2, CYP2A6, CYP3A4, CYP3A5, and UGT2B7. dme 30-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 26779253-4 2015 The drug metabolizing enzyme (DME), CYP2B6, is primarily responsible for EFV metabolism with minor contributions by CYP1A2, CYP2A6, CYP3A4, CYP3A5, and UGT2B7. efavirenz 73-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 26580670-0 2016 Effects of the selected cytochrome P450 oxidoreductase genetic polymorphisms on cytochrome P450 2B6 activity as measured by bupropion hydroxylation. Bupropion 124-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-99 26580670-2 2016 The aim of this study was to investigate whether single-nucleotide polymorphisms in the POR gene were correlated with interindividual variations in CYP2B6 activity, as measured by bupropion hydroxylation. Bupropion 180-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-154 26580670-4 2016 The activity of CYP2B6 was evaluated on the basis of the area under the time-concentration curve (AUC) ratio of hydroxybupropion versus bupropion (AUC_hyd/AUC_bup). hydroxybupropion 112-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 26580670-4 2016 The activity of CYP2B6 was evaluated on the basis of the area under the time-concentration curve (AUC) ratio of hydroxybupropion versus bupropion (AUC_hyd/AUC_bup). Bupropion 119-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 26580670-9 2016 CONCLUSION: These results indicate, for the first time, that the POR g.6593A>G polymorphism significantly influences CYP2B6 activity, as measured by bupropion hydroxylation, in humans, and the CYP2B6*6 and POR g.6593A>G polymorphisms might be considered simultaneously for the individualized therapy with CYP2B6 substrate drugs such as bupropion. Bupropion 152-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 26580670-9 2016 CONCLUSION: These results indicate, for the first time, that the POR g.6593A>G polymorphism significantly influences CYP2B6 activity, as measured by bupropion hydroxylation, in humans, and the CYP2B6*6 and POR g.6593A>G polymorphisms might be considered simultaneously for the individualized therapy with CYP2B6 substrate drugs such as bupropion. Bupropion 342-351 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 26580670-9 2016 CONCLUSION: These results indicate, for the first time, that the POR g.6593A>G polymorphism significantly influences CYP2B6 activity, as measured by bupropion hydroxylation, in humans, and the CYP2B6*6 and POR g.6593A>G polymorphisms might be considered simultaneously for the individualized therapy with CYP2B6 substrate drugs such as bupropion. Bupropion 342-351 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 196-202 26580670-9 2016 CONCLUSION: These results indicate, for the first time, that the POR g.6593A>G polymorphism significantly influences CYP2B6 activity, as measured by bupropion hydroxylation, in humans, and the CYP2B6*6 and POR g.6593A>G polymorphisms might be considered simultaneously for the individualized therapy with CYP2B6 substrate drugs such as bupropion. Bupropion 342-351 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 196-202 27515451-0 2016 CYP2B6 and OPRM1 Receptor Polymorphisms at Methadone Clinics And Novel OPRM1 Haplotypes: A Cross-Sectional Study. Methadone 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 27515451-3 2016 OBJECTIVE: This study sought to detect CYP2B6 and OPRM1 variants and their genotypes, as major contributors to inter-variability in methadone responsiveness and methadone dose requirements. Methadone 132-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 27515451-3 2016 OBJECTIVE: This study sought to detect CYP2B6 and OPRM1 variants and their genotypes, as major contributors to inter-variability in methadone responsiveness and methadone dose requirements. Methadone 161-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 26553012-1 2016 Efavirenz (EFV) is principally metabolized by CYP2B6 to 8-hydroxy-efavirenz (8OH-EFV) and to a lesser extent by CYP2A6 to 7-hydroxy-efavirenz (7OH-EFV). efavirenz 11-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 26553012-1 2016 Efavirenz (EFV) is principally metabolized by CYP2B6 to 8-hydroxy-efavirenz (8OH-EFV) and to a lesser extent by CYP2A6 to 7-hydroxy-efavirenz (7OH-EFV). 8-hydroxyefavirenz 56-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 26553012-1 2016 Efavirenz (EFV) is principally metabolized by CYP2B6 to 8-hydroxy-efavirenz (8OH-EFV) and to a lesser extent by CYP2A6 to 7-hydroxy-efavirenz (7OH-EFV). 8oh-efv 77-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 26553012-6 2016 In individuals (n = 67) genotyped for CYP2B6, 2A6, and CYP3A metabolic pathways, 8OH-EFV/EFV ratios in plasma were an index of CYP2B6 phenotypic activity (P < 0.0001), which was also reflected by phase II metabolites 8OH-EFV-glucuronide/EFV and 8OH-EFV-sulfate/EFV ratios. 2-Phenyl-~{n}4-(Thiophen-2-Ylmethyl)quinazoline-4,7-Diamine 81-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 26553012-6 2016 In individuals (n = 67) genotyped for CYP2B6, 2A6, and CYP3A metabolic pathways, 8OH-EFV/EFV ratios in plasma were an index of CYP2B6 phenotypic activity (P < 0.0001), which was also reflected by phase II metabolites 8OH-EFV-glucuronide/EFV and 8OH-EFV-sulfate/EFV ratios. 2-Phenyl-~{n}4-(Thiophen-2-Ylmethyl)quinazoline-4,7-Diamine 81-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 26553012-6 2016 In individuals (n = 67) genotyped for CYP2B6, 2A6, and CYP3A metabolic pathways, 8OH-EFV/EFV ratios in plasma were an index of CYP2B6 phenotypic activity (P < 0.0001), which was also reflected by phase II metabolites 8OH-EFV-glucuronide/EFV and 8OH-EFV-sulfate/EFV ratios. efavirenz 85-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 26681005-1 2015 CYP2B6 is a highly polymorphic isoenzyme involved in the metabolism of many drugs including cyclophosphamide, bupropion, and efavirenz. Cyclophosphamide 92-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26338349-14 2016 An IPRN-derived furanoepoxide and/or gamma-ketoenal intermediate(s) were/was generated and may be responsible for the inactivation of CYP2B6. furanoepoxide 16-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-140 26338349-14 2016 An IPRN-derived furanoepoxide and/or gamma-ketoenal intermediate(s) were/was generated and may be responsible for the inactivation of CYP2B6. gamma-ketoenal 37-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-140 26681005-1 2015 CYP2B6 is a highly polymorphic isoenzyme involved in the metabolism of many drugs including cyclophosphamide, bupropion, and efavirenz. Bupropion 110-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26681005-1 2015 CYP2B6 is a highly polymorphic isoenzyme involved in the metabolism of many drugs including cyclophosphamide, bupropion, and efavirenz. efavirenz 125-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26335194-0 2015 Psoralen, a mechanism-based inactivator of CYP2B6. Ficusin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 26126958-5 2015 alpha-Naphthoflavone (a selective CYP1A2 inhibitor), thioTEPA (a CYP2B6 inhibitor) and anti-CYP2B6 antibody inhibited (-)-cis-rose oxide 9-hydroxylation catalysed by human liver microsomes. Thiotepa 53-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 26126958-5 2015 alpha-Naphthoflavone (a selective CYP1A2 inhibitor), thioTEPA (a CYP2B6 inhibitor) and anti-CYP2B6 antibody inhibited (-)-cis-rose oxide 9-hydroxylation catalysed by human liver microsomes. cis-rose oxide 118-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 26126958-5 2015 alpha-Naphthoflavone (a selective CYP1A2 inhibitor), thioTEPA (a CYP2B6 inhibitor) and anti-CYP2B6 antibody inhibited (-)-cis-rose oxide 9-hydroxylation catalysed by human liver microsomes. cis-rose oxide 118-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 26126958-6 2015 On the other hand, the metabolism of (-)-trans-rose oxide was suppressed by thioTEPA and anti-CYP2B6 at a significant level in human liver microsomes. trans-Rose oxide 37-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 26126958-8 2015 Using microsomal preparations from nine different human liver samples, (-)-9-hydroxy-cis- and (-)-9-hydroxy-trans-rose oxide formations correlated with (S)-mephenytoin N-demethylase activity (CYP2B6 marker activity). (-)-9-hydroxy-cis- and (-)-9-hydroxy-trans-rose oxide 71-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 26126958-8 2015 Using microsomal preparations from nine different human liver samples, (-)-9-hydroxy-cis- and (-)-9-hydroxy-trans-rose oxide formations correlated with (S)-mephenytoin N-demethylase activity (CYP2B6 marker activity). Mephenytoin 152-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 26126958-9 2015 These results suggest that CYP2B6 plays important roles in the metabolism of (-)-cis- and (-)-trans-rose oxide in human liver microsomes. (-)-cis- and (-)-trans-rose oxide 77-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 26544874-0 2015 Cytochrome P450 Oxidoreductase Influences CYP2B6 Activity in Cyclophosphamide Bioactivation. Cyclophosphamide 61-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 26544874-2 2015 Cyclophosphamide is a prodrug activated mainly by cytochrome P450 2B6 (CYP2B6) in the liver. Cyclophosphamide 0-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-69 26544874-2 2015 Cyclophosphamide is a prodrug activated mainly by cytochrome P450 2B6 (CYP2B6) in the liver. Cyclophosphamide 0-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 26544874-4 2015 MATERIALS AND METHODS: Hydroxylation of cyclophosphamide was investigated in vitro using three microsomal batches of CYP2B6*1 with different ratios of POR/CYP expression levels. Cyclophosphamide 40-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 26544874-14 2015 CONCLUSIONS: This investigation shows for the first time that POR besides CYP2B6 can influence cyclophosphamide metabolism. Cyclophosphamide 95-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 26276083-6 2015 Studies showed the involvement of CYP1A2, CYP2B6, CYP2C19, and CYP3A4 in N-dealkylation of all three compounds and additionally CYP2D6 for lefetamine and NEDPA. Nitrogen 73-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 26276083-6 2015 Studies showed the involvement of CYP1A2, CYP2B6, CYP2C19, and CYP3A4 in N-dealkylation of all three compounds and additionally CYP2D6 for lefetamine and NEDPA. lephetamine 139-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 26126958-1 2015 The in vitro metabolism of (-)-cis- and (-)-trans-rose oxide was investigated using human liver microsomes and recombinant cytochrome P450 (P450 or CYP) enzymes for the first time. (-)-cis- and (-)-trans-rose oxide 27-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-151 26126958-3 2015 Of 11 different recombinant human P450 enzymes used, CYP2B6 and CYP2C19 were the primary enzymes catalysing the metabolism of (-)-cis- and (-)-trans-rose oxide. (-)-cis- and 126-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 26126958-3 2015 Of 11 different recombinant human P450 enzymes used, CYP2B6 and CYP2C19 were the primary enzymes catalysing the metabolism of (-)-cis- and (-)-trans-rose oxide. trans-Rose oxide 139-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 26602960-0 2015 The prognostic values of CYP2B6 genetic polymorphisms and metastatic sites for advanced breast cancer patients treated with docetaxel and thiotepa. Docetaxel 124-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 26602960-0 2015 The prognostic values of CYP2B6 genetic polymorphisms and metastatic sites for advanced breast cancer patients treated with docetaxel and thiotepa. Thiotepa 138-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 26602960-8 2015 Genotypes of CYP2B6 had an interaction with clinical efficacy of docetaxel and thiotepa on metastatic breast cancer patients; and metastatic sites also affected clinical responses. Docetaxel 65-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 26602960-8 2015 Genotypes of CYP2B6 had an interaction with clinical efficacy of docetaxel and thiotepa on metastatic breast cancer patients; and metastatic sites also affected clinical responses. Thiotepa 79-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 26276083-6 2015 Studies showed the involvement of CYP1A2, CYP2B6, CYP2C19, and CYP3A4 in N-dealkylation of all three compounds and additionally CYP2D6 for lefetamine and NEDPA. N-Ethyl-alpha-phenylphenethylamine hydrochloride 154-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 26389554-0 2015 Methadone Pharmacogenetics: CYP2B6 Polymorphisms Determine Plasma Concentrations, Clearance, and Metabolism. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 26389554-2 2015 Cytochrome P4502B6 (CYP2B6) is the principle determinant of clinical methadone elimination. Methadone 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 26389554-4 2015 CYP2B6.6, the protein encoded by the CYP2B6*6 polymorphism, deficiently catalyzes methadone metabolism in vitro. Methadone 82-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26389554-4 2015 CYP2B6.6, the protein encoded by the CYP2B6*6 polymorphism, deficiently catalyzes methadone metabolism in vitro. Methadone 82-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 26389554-5 2015 This investigation determined the influence of CYP2B6*6, and other allelic variants encountered, on methadone concentrations, clearance, and metabolism. Methadone 100-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 26389554-8 2015 RESULTS: Average S-methadone apparent oral clearance was 35 and 45% lower in CYP2B6*1/*6 and CYP2B6*6/*6 genotypes, respectively, compared with CYP2B6*1/*1. Methadone 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 26389554-8 2015 RESULTS: Average S-methadone apparent oral clearance was 35 and 45% lower in CYP2B6*1/*6 and CYP2B6*6/*6 genotypes, respectively, compared with CYP2B6*1/*1. Methadone 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 26389554-8 2015 RESULTS: Average S-methadone apparent oral clearance was 35 and 45% lower in CYP2B6*1/*6 and CYP2B6*6/*6 genotypes, respectively, compared with CYP2B6*1/*1. Methadone 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 26389554-9 2015 R-methadone apparent oral clearance was 25 and 35% lower in CYP2B6*1/*6 and CYP2B6*6/*6 genotypes, respectively, compared with CYP2B6*1/*1. Methadone 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 26389554-9 2015 R-methadone apparent oral clearance was 25 and 35% lower in CYP2B6*1/*6 and CYP2B6*6/*6 genotypes, respectively, compared with CYP2B6*1/*1. Methadone 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 26389554-9 2015 R-methadone apparent oral clearance was 25 and 35% lower in CYP2B6*1/*6 and CYP2B6*6/*6 genotypes, respectively, compared with CYP2B6*1/*1. Methadone 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 26389554-10 2015 R- and S-methadone apparent oral clearance was threefold and fourfold greater in CYP2B6*4 carriers. r- and s-methadone 0-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 26389554-11 2015 IV and oral R- and S-methadone metabolism was significantly lower in CYP2B6*6 carriers compared with that of CYP2B6*1 homozygotes and greater in CYP2B6*4 carriers. r- and s-methadone 12-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 26389554-11 2015 IV and oral R- and S-methadone metabolism was significantly lower in CYP2B6*6 carriers compared with that of CYP2B6*1 homozygotes and greater in CYP2B6*4 carriers. r- and s-methadone 12-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 26389554-11 2015 IV and oral R- and S-methadone metabolism was significantly lower in CYP2B6*6 carriers compared with that of CYP2B6*1 homozygotes and greater in CYP2B6*4 carriers. r- and s-methadone 12-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 26389554-12 2015 Methadone metabolism and clearance were lower in African Americans in part because of the CYP2B6*6 genetic polymorphism. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 26389554-13 2015 CONCLUSIONS: CYP2B6 polymorphisms influence methadone plasma concentrations, because of altered methadone metabolism and thus clearance. Methadone 44-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 26389554-13 2015 CONCLUSIONS: CYP2B6 polymorphisms influence methadone plasma concentrations, because of altered methadone metabolism and thus clearance. Methadone 96-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 26389554-15 2015 CYP2B6 pharmacogenetics explains, in part, interindividual variability in methadone elimination. Methadone 74-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26389554-16 2015 CYP2B6 genetic effects on methadone metabolism and clearance may identify subjects at risk for methadone toxicity and drug interactions. Methadone 26-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26389554-16 2015 CYP2B6 genetic effects on methadone metabolism and clearance may identify subjects at risk for methadone toxicity and drug interactions. Methadone 95-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26307675-0 2015 Application of HC-AFW1 Hepatocarcinoma Cells for Mechanistic Studies: Regulation of Cytochrome P450 2B6 Expression by Dimethyl Sulfoxide and Early Growth Response 1. Dimethyl Sulfoxide 118-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-103 26307675-1 2015 Various exogenous compounds, for example, the drugs bupropione and propofol, but also various cytostatics, are metabolized in the liver by the enzyme cytochrome P450 (P450) CYP2B6. bupropione 52-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 26307675-1 2015 Various exogenous compounds, for example, the drugs bupropione and propofol, but also various cytostatics, are metabolized in the liver by the enzyme cytochrome P450 (P450) CYP2B6. Propofol 67-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 26307675-7 2015 By contrast, CYP2B6 mRNA was strongly induced by DMSO. Dimethyl Sulfoxide 49-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 26307675-11 2015 These findings demonstrate that EGR1 is involved in the regulation of CYP2B6 by DMSO in HC-AFW1 cells. Dimethyl Sulfoxide 80-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 26335194-3 2015 We evaluated the inhibitory effect of PRN on cytochrome P450 2B6 (CYP2B6) and found that PRN induced a time-, concentration-, and NADPH-dependent inactivation of CYP2B6 with the values of KI and kinact being 110.2 muM and 0.200 min(-1), respectively. NADP 130-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-64 26335194-3 2015 We evaluated the inhibitory effect of PRN on cytochrome P450 2B6 (CYP2B6) and found that PRN induced a time-, concentration-, and NADPH-dependent inactivation of CYP2B6 with the values of KI and kinact being 110.2 muM and 0.200 min(-1), respectively. NADP 130-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 26335194-3 2015 We evaluated the inhibitory effect of PRN on cytochrome P450 2B6 (CYP2B6) and found that PRN induced a time-, concentration-, and NADPH-dependent inactivation of CYP2B6 with the values of KI and kinact being 110.2 muM and 0.200 min(-1), respectively. NADP 130-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 26335194-4 2015 Ticlopidine, a CYP2B6 substrate, prevented the enzyme from the inactivation induced by PRN. Ticlopidine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 26335194-5 2015 Exogenous nucleophile glutathione (GSH) and catalase/superoxide dismutase showed limited protection of CYP2B6 from the inactivation. Glutathione 22-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 26335194-5 2015 Exogenous nucleophile glutathione (GSH) and catalase/superoxide dismutase showed limited protection of CYP2B6 from the inactivation. Glutathione 35-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 25565674-0 2015 Effect of CYP2B6*6 on Steady-State Serum Concentrations of Bupropion and Hydroxybupropion in Psychiatric Patients: A Study Based on Therapeutic Drug Monitoring Data. Bupropion 59-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 26220948-6 2015 Kinetic studies using expressed enzymes demonstrated that the contributions of CYP2B6, CYP2C19, and CYP3A4 to the formation of H4 from 2-oxo-clopidogrel were 18.5%, 26.1%, and 53.5%, respectively. 2-oxo-clopidogrel 135-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 26220948-7 2015 The CLint ratios of H3 formation to H4 formation from 2-oxo-clopidogrel by CYP2B6, CYP2C19, and CYP3A4 were 2.2, 1.0, and 1.7, respectively. HS 3 20-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 26220948-7 2015 The CLint ratios of H3 formation to H4 formation from 2-oxo-clopidogrel by CYP2B6, CYP2C19, and CYP3A4 were 2.2, 1.0, and 1.7, respectively. 2-oxo-clopidogrel 54-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 26247717-0 2015 CYP2B6 poor metaboliser alleles involved in efavirenz and nevirapine metabolism: CYP2B6*9 and CYP2B6*18 distribution in HIV-exposed subjects from Dschang, Western Cameroon. Nevirapine 58-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 25601761-8 2015 This is the first study demonstrating that CYP2J2 is equally important to CYP2B6 in Cy metabolism. Cyclophosphamide 84-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 26516577-7 2015 In addition, PXR (rifampicin) and CAR-selective (carbamazepine and phenytoin) inducers of CYP3A4 and CYP2B6 induction, respectively, were demonstrated. Rifampin 18-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 26516577-7 2015 In addition, PXR (rifampicin) and CAR-selective (carbamazepine and phenytoin) inducers of CYP3A4 and CYP2B6 induction, respectively, were demonstrated. Carbamazepine 49-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 26516577-7 2015 In addition, PXR (rifampicin) and CAR-selective (carbamazepine and phenytoin) inducers of CYP3A4 and CYP2B6 induction, respectively, were demonstrated. Phenytoin 67-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 26160115-10 2015 Moreover, by luciferase reporter assay and quantitative real-time PCR analysis, pyrene was found to be a potential inducer of CYP2B6 expression via activation of human CAR in HepG2 cells and human primary hepatocytes. pyrene 80-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 25565674-0 2015 Effect of CYP2B6*6 on Steady-State Serum Concentrations of Bupropion and Hydroxybupropion in Psychiatric Patients: A Study Based on Therapeutic Drug Monitoring Data. hydroxybupropion 73-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 25565674-2 2015 Previous studies have reported conflicting impact of the CYP2B6*6 variant allele on the formation of hydroxybupropion from bupropion. hydroxybupropion 101-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 25565674-2 2015 Previous studies have reported conflicting impact of the CYP2B6*6 variant allele on the formation of hydroxybupropion from bupropion. Bupropion 108-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 25565674-3 2015 The aim of this study was to clarify the effect of CYP2B6*6 and secondarily CYP2D6 genotype on steady-state serum concentrations of bupropion and hydroxybupropion in a large population of psychiatric patients. Bupropion 132-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 25565674-11 2015 CONCLUSIONS: This study shows that the CYP2B6*6 variant allele is associated with significantly reduced formation of the active bupropion metabolite in psychiatric patients. Bupropion 128-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 25565674-12 2015 Our findings suggest that dose-adjusted serum concentrations of hydroxybupropion at steady state is approximately halved in homozygous CYP2B6*6 carriers, which might imply risk of reduced clinical response in this patient subgroup. hydroxybupropion 64-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 26366873-0 2015 Optical Isomers of Atorvastatin, Rosuvastatin and Fluvastatin Enantiospecifically Activate Pregnane X Receptor PXR and Induce CYP2A6, CYP2B6 and CYP3A4 in Human Hepatocytes. Rosuvastatin Calcium 33-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-140 26366873-0 2015 Optical Isomers of Atorvastatin, Rosuvastatin and Fluvastatin Enantiospecifically Activate Pregnane X Receptor PXR and Induce CYP2A6, CYP2B6 and CYP3A4 in Human Hepatocytes. Fluvastatin 50-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-140 26366873-0 2015 Optical Isomers of Atorvastatin, Rosuvastatin and Fluvastatin Enantiospecifically Activate Pregnane X Receptor PXR and Induce CYP2A6, CYP2B6 and CYP3A4 in Human Hepatocytes. Atorvastatin 19-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-140 26404338-2 2015 In addition, the inhibitory potential of TCAS on major CYP450 drug metabolizing enzymes (CYP1A2, CYP2C9, CYP2B6, CYP2D6 and CYP3A4) was assessed. Trichloroacetic Acid 41-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 26153084-0 2015 CYP2B6 rs2279343 polymorphism is associated with smoking cessation success in bupropion therapy. Bupropion 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26336590-9 2015 The results showed that mesalazine promotes CYP2B6 and 3A4 activities, while mosapride citrate promotes CYP1A2, 2B6, and 3A4 activities. Mesalamine 24-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 26153084-1 2015 BACKGROUND: Previous studies suggested that polymorphisms in the CYP2B6 gene (which encodes an isoenzyme that metabolizes bupropion) and in the ANKK1 gene (which is located in the ANKK1/DRD2 gene cluster) might influence response to therapy. Bupropion 122-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 26153084-7 2015 RESULTS: Patients with CYP2B6 rs2279343 wild-type AA genotype had higher success rate (48.0 %) compared with patients carrying AG or GG genotypes (CYP2B6*4 variant) (35.5 %) on bupropion therapy. Bupropion 177-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 26153084-10 2015 CONCLUSION: We showed that patients with CYP2B6*4 (rs2279343) variant had lower success rate with bupropion. Bupropion 98-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 26153084-11 2015 Likely, the CYP2B6*4 variant, which leads to a rapid predicted metabolic phenotype for the isoenzyme, influences the pharmacological activity of bupropion. Bupropion 145-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 26153084-12 2015 Our finding suggests that CYP2B6*4 may be an important genetic marker for individualized bupropion pharmacotherapy. Bupropion 89-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 25776278-10 2015 In conclusion, SIPI5357 might cause drug-drug interaction via induction of CYP2B6. sipi5357 15-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 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 2 subfamily B member 6 Homo sapiens 67-73 25616378-1 2015 Benzo[a]pyrene-7,8-dione (BPQ) is formed by the activation of benzo[a]pyrene(B[a]P), which is an environmental toxic substance that is easily exposed in daily life, due to P450/epoxide hydrolase, and is a substance that induces DNA deformation by forming adducts with DNA. benzo(a)pyrene-7,8-dione 0-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 25616378-1 2015 Benzo[a]pyrene-7,8-dione (BPQ) is formed by the activation of benzo[a]pyrene(B[a]P), which is an environmental toxic substance that is easily exposed in daily life, due to P450/epoxide hydrolase, and is a substance that induces DNA deformation by forming adducts with DNA. benzo(a)pyrene-7,8-dione 26-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 25616378-1 2015 Benzo[a]pyrene-7,8-dione (BPQ) is formed by the activation of benzo[a]pyrene(B[a]P), which is an environmental toxic substance that is easily exposed in daily life, due to P450/epoxide hydrolase, and is a substance that induces DNA deformation by forming adducts with DNA. Benzo(a)pyrene 62-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 26279982-5 2015 For other tests further studies are needed: CYP2B6 516 G > T testing may be able to identify patients at higher risk of Central Nervous System side effects following standard dosing of Efavirenz, UGT1A1*28 testing before initiation of antiretroviral therapy containing Atazanavir may aid in identifying individuals at risk of hyperbilirubinaemia. Atazanavir Sulfate 272-282 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 26135009-4 2015 Desloratadine (10 microM) caused no inhibition (<15%) of CYP1A2, CYP2C8, CYP2C9, or CYP2C19 and weak inhibition (32-48%) of CYP2B6, CYP2D6, and CYP3A4/5. desloratadine 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 26348712-0 2015 Effects of CYP2B6 and CYP1A2 Genetic Variation on Nevirapine Plasma Concentration and Pharmacodynamics as Measured by CD4 Cell Count in Zimbabwean HIV-Infected Patients. Nevirapine 50-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 26348712-4 2015 This study, therefore, aimed to identify sources of variability in nevirapine pharmacokinetics and pharmacodynamics, focusing on genetic variation in CYP2B6 and CYP1A2. Nevirapine 67-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 26348712-7 2015 CYP2B6 and CYP1A2 were chosen as the candidate genes based on reports in literature, as well as their prominence in the metabolism of efavirenz, a drug in the same class with nevirapine. Nevirapine 175-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26348712-13 2015 We conclude that CYP2B6 c.516G>T and CYP2B6 c.983T>C could be important sources of nevirapine pharmacokinetic variability that could be considered for dosage optimization, while CYP1A2 g.-163C>A seems to be associated with HIV disease progression. Nevirapine 89-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 26348712-13 2015 We conclude that CYP2B6 c.516G>T and CYP2B6 c.983T>C could be important sources of nevirapine pharmacokinetic variability that could be considered for dosage optimization, while CYP1A2 g.-163C>A seems to be associated with HIV disease progression. Nevirapine 89-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 25702819-0 2015 CYP2B6*6 allele and age substantially reduce steady-state ketamine clearance in chronic pain patients: impact on adverse effects. Ketamine 58-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 25702819-2 2015 The CYP2B6*6 allele is associated with substantially reduced ketamine metabolism in vitro and, therefore, may affect ketamine clearance. Ketamine 61-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 25702819-2 2015 The CYP2B6*6 allele is associated with substantially reduced ketamine metabolism in vitro and, therefore, may affect ketamine clearance. Ketamine 117-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 25702819-3 2015 Our aims were to examine the impact of the CYP2B6*6 allele on ketamine plasma clearance and on adverse effects in chronic pain patients. Ketamine 62-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 25702819-4 2015 METHODS: CYP2B6 genotypes were identified in 49 chronic pain patients who received 24 h continuous subcutaneous infusions of ketamine. Ketamine 125-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 25702819-6 2015 RESULTS: The median plasma clearance of ketamine after 100 mg 24 h(-1) dose was significantly lower in patients with the CYP2B6*6/*6 (21.6 l h(-1) ) and CYP2B6*1/*6 (40.6 l h(-1) ) genotypes compared with patients with the CYP2B6*1/*1 genotype (68.1 l h(-1) , P < 0.001). Ketamine 40-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-127 25702819-6 2015 RESULTS: The median plasma clearance of ketamine after 100 mg 24 h(-1) dose was significantly lower in patients with the CYP2B6*6/*6 (21.6 l h(-1) ) and CYP2B6*1/*6 (40.6 l h(-1) ) genotypes compared with patients with the CYP2B6*1/*1 genotype (68.1 l h(-1) , P < 0.001). Ketamine 40-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 25702819-6 2015 RESULTS: The median plasma clearance of ketamine after 100 mg 24 h(-1) dose was significantly lower in patients with the CYP2B6*6/*6 (21.6 l h(-1) ) and CYP2B6*1/*6 (40.6 l h(-1) ) genotypes compared with patients with the CYP2B6*1/*1 genotype (68.1 l h(-1) , P < 0.001). Ketamine 40-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 25702819-7 2015 The ketamine : norketamine plasma metabolic ratio was significantly higher in patients with the CYP2B6*6/*6 genotype than in those with the CYP2B6*1/*6 and the CYP2B6*1/*1 genotypes (P < 0.001). Ketamine 4-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 25702819-7 2015 The ketamine : norketamine plasma metabolic ratio was significantly higher in patients with the CYP2B6*6/*6 genotype than in those with the CYP2B6*1/*6 and the CYP2B6*1/*1 genotypes (P < 0.001). Ketamine 4-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 25702819-7 2015 The ketamine : norketamine plasma metabolic ratio was significantly higher in patients with the CYP2B6*6/*6 genotype than in those with the CYP2B6*1/*6 and the CYP2B6*1/*1 genotypes (P < 0.001). Ketamine 4-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 25702819-7 2015 The ketamine : norketamine plasma metabolic ratio was significantly higher in patients with the CYP2B6*6/*6 genotype than in those with the CYP2B6*1/*6 and the CYP2B6*1/*1 genotypes (P < 0.001). norketamine 15-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 25702819-7 2015 The ketamine : norketamine plasma metabolic ratio was significantly higher in patients with the CYP2B6*6/*6 genotype than in those with the CYP2B6*1/*6 and the CYP2B6*1/*1 genotypes (P < 0.001). norketamine 15-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 25702819-7 2015 The ketamine : norketamine plasma metabolic ratio was significantly higher in patients with the CYP2B6*6/*6 genotype than in those with the CYP2B6*1/*6 and the CYP2B6*1/*1 genotypes (P < 0.001). norketamine 15-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 25702819-11 2015 CONCLUSIONS: The CYP2B6*6 allele is associated with a substantial decrease in steady-state ketamine plasma clearance in chronic pain patients. Ketamine 91-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 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 2 subfamily B member 6 Homo sapiens 67-73 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 2 subfamily B member 6 Homo sapiens 67-73 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 26075493-0 2015 Mechanism-Based Inactivation of Human Cytochrome P450 2B6 by Chlorpyrifos. Chlorpyrifos 61-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-57 26075493-3 2015 CYP2B6 was previously determined to have the greatest catalytic efficiency for CPO formation in vitro. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 79-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26075493-6 2015 We further evaluated the ability of other organophosphate pesticides including chorpyrifos-methyl, diazinon, parathion-methyl, and azinophos-methyl to inactivate CYP2B6. Organophosphates 42-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 26075493-6 2015 We further evaluated the ability of other organophosphate pesticides including chorpyrifos-methyl, diazinon, parathion-methyl, and azinophos-methyl to inactivate CYP2B6. chorpyrifos-methyl 79-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 26075493-6 2015 We further evaluated the ability of other organophosphate pesticides including chorpyrifos-methyl, diazinon, parathion-methyl, and azinophos-methyl to inactivate CYP2B6. Diazinon 99-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 26075493-6 2015 We further evaluated the ability of other organophosphate pesticides including chorpyrifos-methyl, diazinon, parathion-methyl, and azinophos-methyl to inactivate CYP2B6. Methyl Parathion 109-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 26075493-6 2015 We further evaluated the ability of other organophosphate pesticides including chorpyrifos-methyl, diazinon, parathion-methyl, and azinophos-methyl to inactivate CYP2B6. Azinphosmethyl 131-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 26075493-7 2015 These organophosphate pesticides were also potent MBIs of CYP2B6 characterized by similar kinact and KI values. Organophosphates 6-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 26075493-8 2015 The inactivation of CYP2B6 by CPS was accompanied by the loss of P450 detectable in the CO reduced spectrum and loss of detectable heme. Heme 131-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 26075493-9 2015 High molecular weight aggregates were observed when inactivated CYP2B6 was run on SDS-PAGE gels indicating protein aggregation. Sodium Dodecyl Sulfate 82-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 26075493-13 2015 These results indicate that CPS and other organophosphate pesticides are potent MBIs of CYP2B6 which may have implications for the toxicity of these pesticides as well as the potential for pesticide-drug interactions. Organophosphates 42-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 25948712-8 2015 Although efavirenz is known as a marker substrate for human CYP2B6, efavirenz was not oxidized by CYP2B6 but by CYP2C9 in monkeys. efavirenz 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 25948712-8 2015 Although efavirenz is known as a marker substrate for human CYP2B6, efavirenz was not oxidized by CYP2B6 but by CYP2C9 in monkeys. efavirenz 68-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25929522-5 2015 Decrease of CYP2A6 and CYP2B6 proteins was observed in rosuvastatin-treated cells. Rosuvastatin Calcium 55-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 25897175-0 2015 Differences in Methadone Metabolism by CYP2B6 Variants. Methadone 15-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 25897175-2 2015 Cytochrome P450 2B6 (CYP2B6) mediates clinical methadone clearance and metabolic inactivation via N-demethylation to 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP). Methadone 47-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 25897175-2 2015 Cytochrome P450 2B6 (CYP2B6) mediates clinical methadone clearance and metabolic inactivation via N-demethylation to 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP). Methadone 47-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 25897175-2 2015 Cytochrome P450 2B6 (CYP2B6) mediates clinical methadone clearance and metabolic inactivation via N-demethylation to 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP). 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine 117-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 25897175-2 2015 Cytochrome P450 2B6 (CYP2B6) mediates clinical methadone clearance and metabolic inactivation via N-demethylation to 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP). 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine 117-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 25897175-2 2015 Cytochrome P450 2B6 (CYP2B6) mediates clinical methadone clearance and metabolic inactivation via N-demethylation to 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP). 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 163-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25897175-2 2015 Cytochrome P450 2B6 (CYP2B6) mediates clinical methadone clearance and metabolic inactivation via N-demethylation to 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP). 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 163-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 25897175-3 2015 Retrospective studies suggest that individuals with the CYP2B6*6 allelic variant have higher methadone plasma concentrations. Methadone 93-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 25897175-5 2015 This investigation evaluated methadone N-demethylation by expressed human CYP2B6 allelic variants in an insect cell coexpression system containing P450 reductase. Methadone 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25839716-0 2015 Genome-wide association study of plasma levels of polychlorinated biphenyls disclose an association with the CYP2B6 gene in a population-based sample. Polychlorinated Biphenyls 50-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. Methadone 54-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 25839716-11 2015 CONCLUSIONS: In our study, we found plasma levels of four lower-chlorinated PCBs to be significantly associated with the genetic region mapping to the CYP2B6 locus. Polychlorinated Biphenyls 76-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 25839716-12 2015 These findings show that CYP2B6 is of importance for the metabolism of PCBs in humans, and may help to identify individuals who may be susceptible to PCB toxicity. Polychlorinated Biphenyls 71-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. Methadone 54-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. Methadone 54-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. Methadone 54-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. Methadone 54-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25897175-7 2015 EDDP formation from therapeutic (0.25-1 muM) R- and S-methadone concentrations was CYP2B6.4 >= CYP2B6.1 >= CYP2B6.5 >> CYP2B6.9 CYP2B6.6, and undetectable from CYP2B6.18. Methadone 54-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25897175-8 2015 Coexpression of b5 had small and variant-specific effects at therapeutic methadone concentrations but at higher concentrations stimulated EDDP formation by CYP2B6.1, CYP2B6.4, CYP2B6.5, and CYP2B6.9 but not CYP2B6.6. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 138-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 156-162 25897175-8 2015 Coexpression of b5 had small and variant-specific effects at therapeutic methadone concentrations but at higher concentrations stimulated EDDP formation by CYP2B6.1, CYP2B6.4, CYP2B6.5, and CYP2B6.9 but not CYP2B6.6. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 138-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 25897175-8 2015 Coexpression of b5 had small and variant-specific effects at therapeutic methadone concentrations but at higher concentrations stimulated EDDP formation by CYP2B6.1, CYP2B6.4, CYP2B6.5, and CYP2B6.9 but not CYP2B6.6. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 138-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 25897175-8 2015 Coexpression of b5 had small and variant-specific effects at therapeutic methadone concentrations but at higher concentrations stimulated EDDP formation by CYP2B6.1, CYP2B6.4, CYP2B6.5, and CYP2B6.9 but not CYP2B6.6. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 138-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 25897175-8 2015 Coexpression of b5 had small and variant-specific effects at therapeutic methadone concentrations but at higher concentrations stimulated EDDP formation by CYP2B6.1, CYP2B6.4, CYP2B6.5, and CYP2B6.9 but not CYP2B6.6. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 138-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 25897175-10 2015 Stereoselective methadone metabolism (S>R) was maintained with all CYP2B6 variants. Methadone 16-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 25897175-11 2015 These results show that methadone N-demethylation by CYP2B6.4 is greater compared with CYP2B6.1, whereas CYP2B6.9 and CYP2B6.6 (which both contain the 516G>T, Q172H polymorphism), are catalytically deficient. Methadone 24-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 25897175-13 2015 Differences in methadone metabolism by CYP2B6 allelic variants provide a mechanistic understanding of pharmacogenetic variability in clinical methadone metabolism and clearance. Methadone 15-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 25897175-13 2015 Differences in methadone metabolism by CYP2B6 allelic variants provide a mechanistic understanding of pharmacogenetic variability in clinical methadone metabolism and clearance. Methadone 142-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 25904761-1 2015 Bupropion"s metabolism and the formation of hydroxybupropion in the liver by cytochrome P450 2B6 (CYP2B6) has been extensively studied; however, the metabolism and formation of erythro/threohydrobupropion in the liver and intestine by carbonyl reductases (CR) has not been well characterized. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-96 25904761-1 2015 Bupropion"s metabolism and the formation of hydroxybupropion in the liver by cytochrome P450 2B6 (CYP2B6) has been extensively studied; however, the metabolism and formation of erythro/threohydrobupropion in the liver and intestine by carbonyl reductases (CR) has not been well characterized. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25904761-1 2015 Bupropion"s metabolism and the formation of hydroxybupropion in the liver by cytochrome P450 2B6 (CYP2B6) has been extensively studied; however, the metabolism and formation of erythro/threohydrobupropion in the liver and intestine by carbonyl reductases (CR) has not been well characterized. hydroxybupropion 44-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-96 26227254-6 2015 CYP2B6 pharmacogenetic testing of methadone may reduce the risk of cardiac toxicity associated with the S-enantiomer. Methadone 34-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 25904761-1 2015 Bupropion"s metabolism and the formation of hydroxybupropion in the liver by cytochrome P450 2B6 (CYP2B6) has been extensively studied; however, the metabolism and formation of erythro/threohydrobupropion in the liver and intestine by carbonyl reductases (CR) has not been well characterized. hydroxybupropion 44-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25904761-1 2015 Bupropion"s metabolism and the formation of hydroxybupropion in the liver by cytochrome P450 2B6 (CYP2B6) has been extensively studied; however, the metabolism and formation of erythro/threohydrobupropion in the liver and intestine by carbonyl reductases (CR) has not been well characterized. erythro/threohydrobupropion 177-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-96 25904761-1 2015 Bupropion"s metabolism and the formation of hydroxybupropion in the liver by cytochrome P450 2B6 (CYP2B6) has been extensively studied; however, the metabolism and formation of erythro/threohydrobupropion in the liver and intestine by carbonyl reductases (CR) has not been well characterized. erythro/threohydrobupropion 177-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 25904761-2 2015 The purpose of this investigation was to compare the relative contribution of the two metabolism pathways of bupropion (by CYP2B6 and CR) in the subcellular fractions of liver and intestine and to identify the CRs responsible for erythro/threohydrobupropion formation in the liver and the intestine. Bupropion 109-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-129 25904761-5 2015 Interestingly, the liver has similar capability to form hydroxybupropion (by CYP2B6) and threohydrobupropion (by CR). hydroxybupropion 56-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 25652250-0 2015 Effect of Brain CYP2B Inhibition on Brain Nicotine Levels and Nicotine Self-Administration. Nicotine 42-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-21 25652250-0 2015 Effect of Brain CYP2B Inhibition on Brain Nicotine Levels and Nicotine Self-Administration. Nicotine 62-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-21 25652250-3 2015 CYP2B can metabolize nicotine, the main psychoactive ingredient in cigarettes; if altered brain CYP2B activity can influence nicotine brain levels, it could influence nicotine-mediated behaviors. Nicotine 21-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 25652250-3 2015 CYP2B can metabolize nicotine, the main psychoactive ingredient in cigarettes; if altered brain CYP2B activity can influence nicotine brain levels, it could influence nicotine-mediated behaviors. Nicotine 21-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-101 25652250-3 2015 CYP2B can metabolize nicotine, the main psychoactive ingredient in cigarettes; if altered brain CYP2B activity can influence nicotine brain levels, it could influence nicotine-mediated behaviors. Nicotine 125-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 25652250-3 2015 CYP2B can metabolize nicotine, the main psychoactive ingredient in cigarettes; if altered brain CYP2B activity can influence nicotine brain levels, it could influence nicotine-mediated behaviors. Nicotine 125-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-101 25652250-3 2015 CYP2B can metabolize nicotine, the main psychoactive ingredient in cigarettes; if altered brain CYP2B activity can influence nicotine brain levels, it could influence nicotine-mediated behaviors. Nicotine 125-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 25652250-3 2015 CYP2B can metabolize nicotine, the main psychoactive ingredient in cigarettes; if altered brain CYP2B activity can influence nicotine brain levels, it could influence nicotine-mediated behaviors. Nicotine 125-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-101 25652250-4 2015 To investigate this, a mechanism-based inhibitor selective for CYP2B, C8-xanthate (20 mug), was administered intracerebroventricularly (ICV) into the brain of rats, and 22 h later, nicotine levels were measured by in vivo microdialysis following nicotine (150 mug/kg intravenous). c8-xanthate 70-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-68 25652250-10 2015 Together these data demonstrate that the brain CYP2B activity can influence nicotine brain levels and subsequent behaviors independent of hepatic metabolism. Nicotine 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-52 25652250-11 2015 This suggests that human smokers with variable CYP2B brain levels could have different nicotine levels and reinforcement, which might have a role in smoking behaviors and dependence. Nicotine 87-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-52 25868557-10 2015 Modeling based on estimated Ki values and plasma concentrations from the literature suggest that the orally administered azoles, particularly ketoconazole and voriconazole, have the greatest potential for inhibiting CYP3A4 pathways, as does voriconazole for the CYP2B6 pathways. Azoles 121-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 262-268 26042637-2 2015 The effectiveness of P450 enzymes as highly selective biocatalysts for a wide range of oxygenation reactions of complex substrates has driven chemists to develop synthetic metalloporphyrin model compounds that mimic P450 reactivity. Metalloporphyrins 172-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 26042637-2 2015 The effectiveness of P450 enzymes as highly selective biocatalysts for a wide range of oxygenation reactions of complex substrates has driven chemists to develop synthetic metalloporphyrin model compounds that mimic P450 reactivity. Metalloporphyrins 172-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 216-220 25882581-4 2015 These metabolites are generated by cytochromes P450 (P450s), as shown by experiments with either N-benzylimidazole as a P450 inhibitor or recombinant human P450s. 1-benzylimidazole 97-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 25882581-4 2015 These metabolites are generated by cytochromes P450 (P450s), as shown by experiments with either N-benzylimidazole as a P450 inhibitor or recombinant human P450s. 1-benzylimidazole 97-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-58 25882581-4 2015 These metabolites are generated by cytochromes P450 (P450s), as shown by experiments with either N-benzylimidazole as a P450 inhibitor or recombinant human P450s. 1-benzylimidazole 97-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 25868557-10 2015 Modeling based on estimated Ki values and plasma concentrations from the literature suggest that the orally administered azoles, particularly ketoconazole and voriconazole, have the greatest potential for inhibiting CYP3A4 pathways, as does voriconazole for the CYP2B6 pathways. Voriconazole 159-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 262-268 25868557-10 2015 Modeling based on estimated Ki values and plasma concentrations from the literature suggest that the orally administered azoles, particularly ketoconazole and voriconazole, have the greatest potential for inhibiting CYP3A4 pathways, as does voriconazole for the CYP2B6 pathways. Ketoconazole 142-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 262-268 26415139-6 2015 Thus, CYP2B6 is a crucial enzyme in the metabolism of antiretroviral drugs, efavirenz and nevirapine. efavirenz 76-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 24664476-12 2015 For all patients, predisposing factors for ifosfamide-induced encephalopathy included previous cisplatin exposure, concomitant opioids and CYP2B6 inhibitors. Ifosfamide 43-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 139-145 26415139-6 2015 Thus, CYP2B6 is a crucial enzyme in the metabolism of antiretroviral drugs, efavirenz and nevirapine. Nevirapine 90-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 25801005-3 2015 Methadone is N-demethylated to 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) by CYP2B6 and CYP3A4 in vitro, but by CYP2B6 in vivo. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 26055126-0 2015 Selective inhibitory effects of machilin A isolated from Machilus thunbergii on human cytochrome P450 1A and 2B6. machilin A 32-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-112 26055126-8 2015 RESULTS: MA strongly inhibited CYP1A2-mediated phenacetin O-deethylation and CYP2B6-mediated bupropion hydroxylation with IC50 values of 3.0 and 3.9 microM, respectively, while it did not significantly inhibit other CYPs. Bupropion 93-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 25765475-3 2015 Non-specific binding and the activity of the enzymes involved in permethrin"s metabolism (cytochromes P450 and carboxylesterases) were quantified. Permethrin 65-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-128 25404103-2 2015 They play important roles in insecticide metabolism and resistance, and growing numbers of P450 enzymes are now known to catalyse important physiological reactions, such as hormone metabolism or cuticular hydrocarbon synthesis. Hydrocarbons 205-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 25860621-4 2015 Experiments using pan- or isoform-selective CYP inhibitors showed that CYP2B6 and CYP3A4 are responsible for the bioactivation of cyclophosphamide. Cyclophosphamide 130-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 25817938-0 2015 Inhibition of cytochrome P450 2B4 by environmentally persistent free radical-containing particulate matter. Free Radicals 64-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 25817938-9 2015 In this study, the inhibition of P450 by MCP230 was examined in more detail by measuring its effect on the rate of metabolism of 7-ethoxy-4-trifluoromethylcoumarin (7EFC) and 7-benzyloxyresorufin (7BRF) by the purified, reconstituted CYP2B4 system. 7-ethoxy-4-trifluoromethylcoumarin 129-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 25801005-3 2015 Methadone is N-demethylated to 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) by CYP2B6 and CYP3A4 in vitro, but by CYP2B6 in vivo. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 25801005-3 2015 Methadone is N-demethylated to 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) by CYP2B6 and CYP3A4 in vitro, but by CYP2B6 in vivo. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 31-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 25817938-9 2015 In this study, the inhibition of P450 by MCP230 was examined in more detail by measuring its effect on the rate of metabolism of 7-ethoxy-4-trifluoromethylcoumarin (7EFC) and 7-benzyloxyresorufin (7BRF) by the purified, reconstituted CYP2B4 system. benzyloxyresorufin 175-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 25801005-3 2015 Methadone is N-demethylated to 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) by CYP2B6 and CYP3A4 in vitro, but by CYP2B6 in vivo. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 82-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 25817938-9 2015 In this study, the inhibition of P450 by MCP230 was examined in more detail by measuring its effect on the rate of metabolism of 7-ethoxy-4-trifluoromethylcoumarin (7EFC) and 7-benzyloxyresorufin (7BRF) by the purified, reconstituted CYP2B4 system. benzyloxyresorufin 197-201 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 25801005-3 2015 Methadone is N-demethylated to 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) by CYP2B6 and CYP3A4 in vitro, but by CYP2B6 in vivo. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 82-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 25801005-8 2015 CYP2B6 mRNA and activity (bupropion N-demethylation) were induced by several antiretrovirals, as were CYP3A4 mRNA and protein expression, but only indinavir increased CYP3A activity (alfentanil dealkylation). Bupropion 26-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 25801005-8 2015 CYP2B6 mRNA and activity (bupropion N-demethylation) were induced by several antiretrovirals, as were CYP3A4 mRNA and protein expression, but only indinavir increased CYP3A activity (alfentanil dealkylation). Alfentanil 183-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 26288843-4 2015 METHODS: We genotyped CYP2A6, CYP2B6 and CYP3A4, which encode enzymes principally involved in EFV metabolism, from patients enrolled in the multinational SMART, FIRST and ESPRIT studies, for whom outcome data of treatment adherence was available. efavirenz 94-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 26288843-8 2015 FINDINGS: Patients with highest pharmacogenetic risk, as defined by cumulative SNPs in CYP2A6, CYP2B6 and CYP3A4, have an increased risk of discontinuation of EFV containing therapy compared to patients with lower genetic risk scores (adjusted HR 1.9, 95% CI 1.2, 3.1, P = 0.009). efavirenz 159-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-101 25906774-2 2015 EFV, NVP, artemether and lumefantrine are substrates, inhibitors or inducers of CYP3A4 and CYP2B6, creating a potential for drug-drug interactions. efavirenz 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 25677220-9 2015 Our results suggest that CYP2B6 SNPs influence the efficacy of high-dose Cy and significantly reduce the success of autologous HCT for lymphoma patients with the CYP2B6*5 variant. Cyclophosphamide 73-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 25677220-9 2015 Our results suggest that CYP2B6 SNPs influence the efficacy of high-dose Cy and significantly reduce the success of autologous HCT for lymphoma patients with the CYP2B6*5 variant. Cyclophosphamide 73-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 25677220-0 2015 Cytochrome P450 2B6*5 Increases Relapse after Cyclophosphamide-Containing Conditioning and Autologous Transplantation for Lymphoma. Cyclophosphamide 46-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 25725071-7 2015 Interestingly, GW4064 did not repress expression of CYP2B6, another target gene of PXR and CAR; GW4064 enhanced CYP2B6 promoter activity. GW 4064 96-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-118 25634537-5 2015 In the model, we tested the hypothesis that metabolism responsibilities were shared by the p450 CYP2E1 and glutathione (GSH) conjugation. Glutathione 120-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 25906774-2 2015 EFV, NVP, artemether and lumefantrine are substrates, inhibitors or inducers of CYP3A4 and CYP2B6, creating a potential for drug-drug interactions. Artemether 10-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 25634537-7 2015 Furthermore, the above model was tested by simulating the gas-uptake data of the female rats pretreated with 1-aminobenzotrizole, a general P450 suicide inhibitor, or d,l-buthionine (S,R)-sulfoximine, an inhibitor of GSH synthesis, prior to exposure to 800 ppm 1-BP. 1-aminobenzotrizole 109-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-144 25906774-2 2015 EFV, NVP, artemether and lumefantrine are substrates, inhibitors or inducers of CYP3A4 and CYP2B6, creating a potential for drug-drug interactions. Lumefantrine 25-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 25634537-8 2015 The comparative investigation on the metabolic pathway of 1-BP through the PBPK modeling in both sexes provides critical information for understanding the role of p450 and GSH in the metabolism of 1-BP and eventually helps to quantitatively extrapolate current animal studies to human. 1-bromopropane 58-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-167 25634537-8 2015 The comparative investigation on the metabolic pathway of 1-BP through the PBPK modeling in both sexes provides critical information for understanding the role of p450 and GSH in the metabolism of 1-BP and eventually helps to quantitatively extrapolate current animal studies to human. 1-bromopropane 197-201 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-167 25629761-0 2015 Primary role of cytochrome P450 2B6 in the oxidative metabolism of 2,2",4,4",6-pentabromodiphenyl ether (BDE-100) to hydroxylated BDEs. pentabromodiphenyl ether 67-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-35 25878720-1 2015 BACKGROUND: Polymorphisms in cytochrome P450 2B6 (CYP2B6) affect the steady state plasma concentration of nevirapine. Nevirapine 106-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-48 25878720-1 2015 BACKGROUND: Polymorphisms in cytochrome P450 2B6 (CYP2B6) affect the steady state plasma concentration of nevirapine. Nevirapine 106-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 25878720-10 2015 RESULTS: Women with CYP2B6 516TT genotype (n=9) had higher mean nevirapine plasma levels (14.33 mug/mL) compared to those with heterozygous 516GT (9.18 mug/mL; n=25) and wild- type 516GG (7.95 mug/mL; n=32) genotypes (P=0.01). Nevirapine 64-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 25878720-11 2015 Women heterozygous for the CYP2B6 983TC genotype (n=13) had higher mean nevirapine plasma levels (12.94 mug/mL), compared to women with the homozygous 983TT (8.35 mug/mL; n=53) genotype (P=0.007). Nevirapine 72-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 25878720-15 2015 CONCLUSIONS: CYP2B6 516G>T and CYP2B6 983T>C genotypes were strongly associated with plasma nevirapine concentration, which predicted immunologic response in women on nevirapine-based antiretroviral therapy. Nevirapine 98-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 25878720-15 2015 CONCLUSIONS: CYP2B6 516G>T and CYP2B6 983T>C genotypes were strongly associated with plasma nevirapine concentration, which predicted immunologic response in women on nevirapine-based antiretroviral therapy. Nevirapine 98-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 25449227-2 2015 In vitro studies indicated that faldaprevir inhibited CYP2B6, CYP2C9, and CYP3A, and was a weak-to-moderate inactivator of CYP3A4. faldaprevir 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 25683389-14 2015 As a result, high amounts of acetaldehyde will circulate for longer time in the blood, until the liver CYP2E1(p450) enzyme system finally metabilizes the acetaldehyde, during that period of time the patients will experience a flushing as well as the people with the "Asian flushing syndrome" suffer when they drink ethanol. Acetaldehyde 29-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-114 25683389-14 2015 As a result, high amounts of acetaldehyde will circulate for longer time in the blood, until the liver CYP2E1(p450) enzyme system finally metabilizes the acetaldehyde, during that period of time the patients will experience a flushing as well as the people with the "Asian flushing syndrome" suffer when they drink ethanol. Acetaldehyde 154-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-114 25683389-14 2015 As a result, high amounts of acetaldehyde will circulate for longer time in the blood, until the liver CYP2E1(p450) enzyme system finally metabilizes the acetaldehyde, during that period of time the patients will experience a flushing as well as the people with the "Asian flushing syndrome" suffer when they drink ethanol. Ethanol 315-322 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-114 25231529-2 2015 Experimental and human studies implicate the vasoconstrictor P450 eicosanoid 20-hydroxyeicosatetraenoic acid (20-HETE) in the pathogenesis of DCI. Eicosanoids 66-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 25585967-0 2015 Structural and biophysical characterization of human cytochromes P450 2B6 and 2A6 bound to volatile hydrocarbons: analysis and comparison. Hydrocarbons 100-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-81 25585967-1 2015 X-ray crystal structures of complexes of cytochromes CYP2B6 and CYP2A6 with the monoterpene sabinene revealed two distinct binding modes in the active sites. monoterpene sabinene 80-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 25585967-2 2015 In CYP2B6, sabinene positioned itself with the putative oxidation site located closer to the heme iron. sabinene 11-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 3-9 25585967-2 2015 In CYP2B6, sabinene positioned itself with the putative oxidation site located closer to the heme iron. Heme 93-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 3-9 25585967-4 2015 Furthermore, results from isothermal titration calorimetry indicated a much more substantial contribution of favorable enthalpy to sabinene binding to CYP2B6 as opposed to CYP2A6, consistent with the previous observations with (+)-alpha-pinene. sabinene 131-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 25585967-5 2015 Structural analysis of CYP2B6 complexes with sabinene and the structurally similar (3)-carene and comparison with previously solved structures revealed how the movement of the F206 side chain influences the volume of the binding pocket. sabinene 45-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 25585967-5 2015 Structural analysis of CYP2B6 complexes with sabinene and the structurally similar (3)-carene and comparison with previously solved structures revealed how the movement of the F206 side chain influences the volume of the binding pocket. 3-carene 83-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 25585967-8 2015 Overall, the findings from multiple techniques illustrate how drugs metabolizing CYP2B6 and CYP2A6 handle a common hydrocarbon found in the environment. Hydrocarbons 115-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 25561246-9 2015 A contribution of CYP2B6 in testosterone metabolism was only found in human and equine microsomes. Testosterone 28-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 25231529-2 2015 Experimental and human studies implicate the vasoconstrictor P450 eicosanoid 20-hydroxyeicosatetraenoic acid (20-HETE) in the pathogenesis of DCI. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 77-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 25231529-2 2015 Experimental and human studies implicate the vasoconstrictor P450 eicosanoid 20-hydroxyeicosatetraenoic acid (20-HETE) in the pathogenesis of DCI. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 110-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 25231529-2 2015 Experimental and human studies implicate the vasoconstrictor P450 eicosanoid 20-hydroxyeicosatetraenoic acid (20-HETE) in the pathogenesis of DCI. dci 142-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 25231529-4 2015 METHODS: Using mass spectrometry, we measured P450 eicosanoids in cerebrospinal fluid (CSF) from 34 SAH patients from 1 to 14 days after admission. Eicosanoids 51-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-50 25231529-10 2015 CONCLUSIONS: Our findings suggest that P450 eicosanoids play an important role in the pathogenesis of DCI. Eicosanoids 44-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 25889207-8 2015 RESULTS: CYP2B6*6 and *18 variant alleles, weight and sex were the most significant covariates explaining 55% of inter-individual variability in EFV clearance. efavirenz 145-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 25721668-4 2015 Using tetracycline inducible-hCAR system in HepG2 cells, we showed that knockdown of PRMT5 with small interfering RNA suppressed tetracycline -induced mRNA expression of CYP2B6 but not of CYP2C9 or CYP3A4. Tetracycline 6-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-176 25560051-3 2015 Tramadol is metabolized to M1 mainly by cytochrome P450 (CYP)2D6 enzyme and to other metabolites by CYP3A4 and CYP2B6. Tramadol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 25721668-4 2015 Using tetracycline inducible-hCAR system in HepG2 cells, we showed that knockdown of PRMT5 with small interfering RNA suppressed tetracycline -induced mRNA expression of CYP2B6 but not of CYP2C9 or CYP3A4. Tetracycline 129-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-176 25656918-0 2015 Evaluation of metabolism dependent inhibition of CYP2B6 mediated bupropion hydroxylation in human liver microsomes by monoamine oxidase inhibitors and prediction of potential as perpetrators of drug interaction. Bupropion 65-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 25656918-1 2015 The objective of the study was to evaluate the metabolism dependent inhibition of CYP2B6 catalyzed bupropion hydroxylation in human liver microsomes by monoamine oxidase (MAO) inhibitors and to predict the drug-drug interaction potential of monoamine oxidase inhibitors as perpetrators of drug interaction. Bupropion 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 25656918-2 2015 Human liver microsomal CYP2B6 activities were investigated using bupropion hydroxylation as probe substrate marker. Bupropion 65-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 25656918-3 2015 The results from single point time dependent inhibition and shift assays suggest that clorgyline, pargyline, phenelzine, and selegiline were metabolism based inhibitors of CYP2B6. Clorgyline 86-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-178 25656918-3 2015 The results from single point time dependent inhibition and shift assays suggest that clorgyline, pargyline, phenelzine, and selegiline were metabolism based inhibitors of CYP2B6. Pargyline 98-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-178 25656918-3 2015 The results from single point time dependent inhibition and shift assays suggest that clorgyline, pargyline, phenelzine, and selegiline were metabolism based inhibitors of CYP2B6. Phenelzine 109-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-178 25656918-3 2015 The results from single point time dependent inhibition and shift assays suggest that clorgyline, pargyline, phenelzine, and selegiline were metabolism based inhibitors of CYP2B6. Selegiline 125-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-178 25656918-4 2015 In IC50 shift assays, clorgyline, pargyline, phenelzine and selegiline are metabolism based inhibitors of CYP2B6 with fold shit of 3.0-, 3.7-, 2.9-, and 11.4-fold respectively. Clorgyline 22-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 25656918-4 2015 In IC50 shift assays, clorgyline, pargyline, phenelzine and selegiline are metabolism based inhibitors of CYP2B6 with fold shit of 3.0-, 3.7-, 2.9-, and 11.4-fold respectively. Pargyline 34-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 25656918-4 2015 In IC50 shift assays, clorgyline, pargyline, phenelzine and selegiline are metabolism based inhibitors of CYP2B6 with fold shit of 3.0-, 3.7-, 2.9-, and 11.4-fold respectively. Phenelzine 45-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 25656918-4 2015 In IC50 shift assays, clorgyline, pargyline, phenelzine and selegiline are metabolism based inhibitors of CYP2B6 with fold shit of 3.0-, 3.7-, 2.9-, and 11.4-fold respectively. Selegiline 60-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 25656918-6 2015 Phenelzine inactivated CYP2B6 with KI and k(inact) values of 44.9 +- 6.9 muM and 0.085 +- 0.003 min(-1) respectively. Phenelzine 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 25656918-12 2015 The likelihood of drug interaction when selegiline co-administered with CYP2B6 substrates is remote. Selegiline 40-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 25656918-13 2015 Caution is required while co-administering phenelzine with substrates that are exclusively metabolized by CYP2B6 enzyme and substrates that have narrow therapeutic index. Phenelzine 43-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 24445680-5 2015 Western blot data indicated that matrine at 140 mg/L at 72 h induced protein expression of CYP2A6, CYP2B6 and CYP3A4. matrine 33-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 25364857-11 2015 These results suggest that cynomolgus monkeys could be a good model for humans, especially with particular characteristics in reduced CLs of some human P450 substrates by aging. Chlorine 134-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-156 25480923-7 2015 Enzyme activities of CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, and CYP3A4 were reduced upon 48-72 hours exposure to IL-6 in PHH and HepaRG. 4,5,6,7-tetrachlorophthalide 123-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 25052959-3 2015 HS-23 slightly inhibited CYP2A6, CYP2B6, CYP2C9, CYP2C19, and CYP3A4 enzyme activities in human liver microsomes with IC50 values of 80.6, 160.7, 169.5, 85.4, and 76.6 mug/mL, respectively. hs-23 0-5 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 25409894-0 2015 Characterization of CYP2B6 in a CYP2B6-humanized mouse model: inducibility in the liver by phenobarbital and dexamethasone and role in nicotine metabolism in vivo. Phenobarbital 91-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 25480923-7 2015 Enzyme activities of CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, and CYP3A4 were reduced upon 48-72 hours exposure to IL-6 in PHH and HepaRG. heparg 131-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 25409894-0 2015 Characterization of CYP2B6 in a CYP2B6-humanized mouse model: inducibility in the liver by phenobarbital and dexamethasone and role in nicotine metabolism in vivo. Dexamethasone 109-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 25409894-0 2015 Characterization of CYP2B6 in a CYP2B6-humanized mouse model: inducibility in the liver by phenobarbital and dexamethasone and role in nicotine metabolism in vivo. Nicotine 135-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 25760540-1 2015 Repeated pretreatment with the antimalarial drug artemisinin (QHS) could lead to reduced exposure to the parent drug, which is mainly mediated by auto-induction of CYP2B6 activity. artemisinin 49-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-170 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Phenobarbital 30-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Phenobarbital 45-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Phenobarbital 45-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Phenobarbital 45-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Dexamethasone 53-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Dexamethasone 53-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Dexamethasone 53-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Dexamethasone 68-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Dexamethasone 68-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 25183402-11 2015 Structure-activity relationship analysis showed that CYP2B6 and CYP3A4 inducers are bulky lipophilic molecules with a higher number of heavy atoms and a lower number of hydrogen bond donors. Hydrogen 169-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 25409894-2 2015 The inducibility of CYP2B6 by phenobarbital (PB) and dexamethasone (DEX), known inducers of CYP2B6 in human liver, was examined in the TG mice, as well as in TG/Cyp2abfgs-null (or "CYP2B6-humanized") mice. Dexamethasone 68-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 25409894-3 2015 Hepatic expression of CYP2B6 mRNA and protein was greatly induced by PB or DEX treatment in both TG and TG/Cyp2abfgs-null mice. Phenobarbital 69-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 25409894-3 2015 Hepatic expression of CYP2B6 mRNA and protein was greatly induced by PB or DEX treatment in both TG and TG/Cyp2abfgs-null mice. Dexamethasone 75-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 25409894-4 2015 Function of the transgenic CYP2B6 was first studied using bupropion as a probe substrate. Bupropion 58-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 25409894-5 2015 In PB-treated mice, the rates of hepatic microsomal hydroxybupropion formation (at 50 muM bupropion) were >4-fold higher in TG/Cyp2abfgs-null than in Cyp2abfgs-null mice (for both male and female mice); the rate difference was accompanied by a 5-fold higher catalytic efficiency in the TG/Cyp2abfgs-null mice and was abolished by an antibody to CYP2B6. Phenobarbital 3-5 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 348-354 25409894-6 2015 The ability of CYP2B6 to metabolize nicotine was then examined, both in vitro and in vivo. Nicotine 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 25409894-9 2015 Thus, the transgenic CYP2B6 was inducible and functional, and, in the absence of mouse CYP2A and CYP2B enzymes, it contributed to nicotine metabolism in vivo. Nicotine 130-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 25489907-0 2015 CYP2B6 gene single-nucleotide polymorphisms in an Italian population sample and relationship with nicotine dependence. Nicotine 98-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 25489907-1 2015 The extensively polymorphic CYP2B6 gene metabolizes endogenous and exogenous compounds, among which are nicotine and bupropion, although its contribution to the systemic metabolism of nicotine still remains controversial. Nicotine 104-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 25489907-1 2015 The extensively polymorphic CYP2B6 gene metabolizes endogenous and exogenous compounds, among which are nicotine and bupropion, although its contribution to the systemic metabolism of nicotine still remains controversial. Bupropion 117-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 25489907-1 2015 The extensively polymorphic CYP2B6 gene metabolizes endogenous and exogenous compounds, among which are nicotine and bupropion, although its contribution to the systemic metabolism of nicotine still remains controversial. Nicotine 184-192 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 25489907-4 2015 The reduced activity of the CYP2B6*6 variant was significantly (p=0.025) distributed among the nicotine-dependent individuals compared to non-nicotine dependents. Nicotine 95-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 25489907-4 2015 The reduced activity of the CYP2B6*6 variant was significantly (p=0.025) distributed among the nicotine-dependent individuals compared to non-nicotine dependents. Nicotine 142-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 25489907-5 2015 Also, the CYP2B6*1/*6 genotype achieved statistical significance (p=0.016) within the nicotine-dependent individuals. Nicotine 86-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 25489907-6 2015 The high occurrence of CYP2B6*6 carriers among nicotine-dependent individuals may suggest a possible involvement in nicotine dependence, with a potential impact on smoking cessation treatments tailored to the individual smoker"s genotype. Nicotine 47-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 25489907-6 2015 The high occurrence of CYP2B6*6 carriers among nicotine-dependent individuals may suggest a possible involvement in nicotine dependence, with a potential impact on smoking cessation treatments tailored to the individual smoker"s genotype. Nicotine 116-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 25500267-9 2015 S-Mephenytoin, a substrate of CYP2B6, mildly prevented the enzyme from the inactivation induced by IIMP. Mephenytoin 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 25462116-6 2015 A highly active P450 BM3 mutant (CYP102A1M11H) was subsequently used for bioactivation of acetaminophen, clozapine, diclofenac (DF) and mefenamic acid (MFA), but hGST P1-1 adducts were only observed for the latter two drugs. Acetaminophen 90-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 26002731-1 2015 Cytochrome P450 (P450 or CYP) catalysis involves the oxygenation of organic compounds via a series of catalytic intermediates, namely, the ferric-peroxo, ferric-hydroperoxo, Compound I (Cpd I) and FeIII-(H2O2) intermediates. ferric-peroxo 139-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-28 25462116-6 2015 A highly active P450 BM3 mutant (CYP102A1M11H) was subsequently used for bioactivation of acetaminophen, clozapine, diclofenac (DF) and mefenamic acid (MFA), but hGST P1-1 adducts were only observed for the latter two drugs. Clozapine 105-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 25462116-6 2015 A highly active P450 BM3 mutant (CYP102A1M11H) was subsequently used for bioactivation of acetaminophen, clozapine, diclofenac (DF) and mefenamic acid (MFA), but hGST P1-1 adducts were only observed for the latter two drugs. Diclofenac 116-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 25462116-6 2015 A highly active P450 BM3 mutant (CYP102A1M11H) was subsequently used for bioactivation of acetaminophen, clozapine, diclofenac (DF) and mefenamic acid (MFA), but hGST P1-1 adducts were only observed for the latter two drugs. Mefenamic Acid 136-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 25462116-6 2015 A highly active P450 BM3 mutant (CYP102A1M11H) was subsequently used for bioactivation of acetaminophen, clozapine, diclofenac (DF) and mefenamic acid (MFA), but hGST P1-1 adducts were only observed for the latter two drugs. Mefenamic Acid 152-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 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 2 subfamily B member 6 Homo sapiens 138-144 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 2 subfamily B member 6 Homo sapiens 138-144 25069801-6 2015 AITC-mediated reduction in the transcriptional activity of PXR and CAR correlated well with the suppression of CYP3A4 and CYP2B6 expression in HepG2 cells, which reflected the reduced catalytic activities of both of these genes following AITC treatment in differentiated HepaRG cells. allyl isothiocyanate 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 26002731-1 2015 Cytochrome P450 (P450 or CYP) catalysis involves the oxygenation of organic compounds via a series of catalytic intermediates, namely, the ferric-peroxo, ferric-hydroperoxo, Compound I (Cpd I) and FeIII-(H2O2) intermediates. ferric-hydroperoxo 154-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-28 26002731-1 2015 Cytochrome P450 (P450 or CYP) catalysis involves the oxygenation of organic compounds via a series of catalytic intermediates, namely, the ferric-peroxo, ferric-hydroperoxo, Compound I (Cpd I) and FeIII-(H2O2) intermediates. Hydrogen Peroxide 204-208 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-28 26002734-5 2015 In the first example, 2,2",4,4"-tetrabromodiphenyl ether (BDE-47), a halogenated aromatic environmental contaminant, was oxidatively biotransformed by human CYP2B6. 2,2',4,4'-tetrabromodiphenyl ether 22-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 26002734-5 2015 In the first example, 2,2",4,4"-tetrabromodiphenyl ether (BDE-47), a halogenated aromatic environmental contaminant, was oxidatively biotransformed by human CYP2B6. 2,2',4,4'-tetrabromodiphenyl ether 58-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 26002734-6 2015 Nine different metabolites of BDE-47 were produced by CYP2B6 via monooxygenase reactions that included aromatic hydroxylation, with and without an NIH-shift, dealkylation and debromination. 2,2',4,4'-tetrabromodiphenyl ether 30-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 26002738-2 2015 P450 enzymes play an important role in the detoxification of exogenous bioactive compounds and hydrophobic xenobiotics (e.g. carcinogens, drugs, environment pollutants, food supplements, medicines, plant products) and in the biotransformation of endogenous bioactive compounds (e.g. amino acids, cholesterol, eicosanoids, saturated/unsaturated fatty acids, melatonin, steroid hormones). Cholesterol 296-307 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 26002738-2 2015 P450 enzymes play an important role in the detoxification of exogenous bioactive compounds and hydrophobic xenobiotics (e.g. carcinogens, drugs, environment pollutants, food supplements, medicines, plant products) and in the biotransformation of endogenous bioactive compounds (e.g. amino acids, cholesterol, eicosanoids, saturated/unsaturated fatty acids, melatonin, steroid hormones). Eicosanoids 309-320 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 26002738-2 2015 P450 enzymes play an important role in the detoxification of exogenous bioactive compounds and hydrophobic xenobiotics (e.g. carcinogens, drugs, environment pollutants, food supplements, medicines, plant products) and in the biotransformation of endogenous bioactive compounds (e.g. amino acids, cholesterol, eicosanoids, saturated/unsaturated fatty acids, melatonin, steroid hormones). saturated/unsaturated fatty acids 322-355 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 26002738-2 2015 P450 enzymes play an important role in the detoxification of exogenous bioactive compounds and hydrophobic xenobiotics (e.g. carcinogens, drugs, environment pollutants, food supplements, medicines, plant products) and in the biotransformation of endogenous bioactive compounds (e.g. amino acids, cholesterol, eicosanoids, saturated/unsaturated fatty acids, melatonin, steroid hormones). Melatonin 357-366 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 26002738-2 2015 P450 enzymes play an important role in the detoxification of exogenous bioactive compounds and hydrophobic xenobiotics (e.g. carcinogens, drugs, environment pollutants, food supplements, medicines, plant products) and in the biotransformation of endogenous bioactive compounds (e.g. amino acids, cholesterol, eicosanoids, saturated/unsaturated fatty acids, melatonin, steroid hormones). Steroids 368-384 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 26002739-10 2015 Thus, creation of hybrid systems by fusion of the modified heme domain of P450s with proteinaceous electron carriers helps obviate the tedious reconstitution procedure and induces novel activities. Heme 59-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-79 26094899-9 2015 These results suggest that clofazimine and prothionamide are likely to cause clinically relevant DDIs when co-administered with products metabolized by CYP3A4 and CYP2B6, respectively. Clofazimine 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 26094899-9 2015 These results suggest that clofazimine and prothionamide are likely to cause clinically relevant DDIs when co-administered with products metabolized by CYP3A4 and CYP2B6, respectively. Prothionamide 43-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 25428516-0 2015 Effects of CYP2B6 genetic polymorphisms in patients receiving cyclophosphamide combination chemotherapy for breast cancer. Cyclophosphamide 62-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 25428516-1 2015 PURPOSE: The purpose of this study was to measure the frequency of three CYP2B6 [CYP2B6*4 (rs2279343), CYP2B6*5 (rs3211371) and CYP2B6*9 (rs3745274)] alleles in patients with breast cancer receiving cyclophosphamide (CP) therapy and test whether these variants are predictors of CP-associated toxicity and efficacy. Cyclophosphamide 199-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 25655887-2 2015 Nicotine, the major psychoactive compound in cigarette smoke, is metabolized by a number of enzymes, including CYP2A6, CYP2B6, FMOs, and UGTs, among others. Nicotine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 25378064-6 2015 The loss of CYP2B6 activity required the presence of NADPH. NADP 53-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 25600204-2 2015 To evaluate time-dependent cytochrome P450 induction precisely, induction of CYP1A2, CYP2B6, and CYP3A4 mRNA was confirmed (>2-fold) by the treatment with omeprazole, phenobarbital, and rifampicin, respectively, for 24 or 48 h on day 3 from the start of culture. Rifampin 189-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 25600204-2 2015 To evaluate time-dependent cytochrome P450 induction precisely, induction of CYP1A2, CYP2B6, and CYP3A4 mRNA was confirmed (>2-fold) by the treatment with omeprazole, phenobarbital, and rifampicin, respectively, for 24 or 48 h on day 3 from the start of culture. Omeprazole 158-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 25600204-2 2015 To evaluate time-dependent cytochrome P450 induction precisely, induction of CYP1A2, CYP2B6, and CYP3A4 mRNA was confirmed (>2-fold) by the treatment with omeprazole, phenobarbital, and rifampicin, respectively, for 24 or 48 h on day 3 from the start of culture. Phenobarbital 170-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 25378064-8 2015 Ticlopidine, a substrate of CYP2B6, showed protection of the enzyme against the inactivation induced by IMP. Ticlopidine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 25264242-3 2014 Human liver microsomal studies indicated that orteronel weakly inhibits CYP1A2, 2C8, 2C9 and 2C19, with IC50 values of 17.8, 27.7, 30.8 and 38.8 microm, respectively, whereas orteronel does not inhibit CYP2B6, 2D6 or 3A4/5 (IC50 > 100 microm). orteronel 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 202-208 25498968-4 2015 Electron transfer by the NADPH-P450 oxidoreductase is required for reduction of the heme of P450, necessary for binding of molecular oxygen. Heme 84-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 25498968-4 2015 Electron transfer by the NADPH-P450 oxidoreductase is required for reduction of the heme of P450, necessary for binding of molecular oxygen. Heme 84-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 25498968-4 2015 Electron transfer by the NADPH-P450 oxidoreductase is required for reduction of the heme of P450, necessary for binding of molecular oxygen. Oxygen 133-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 25498968-4 2015 Electron transfer by the NADPH-P450 oxidoreductase is required for reduction of the heme of P450, necessary for binding of molecular oxygen. Oxygen 133-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 25316200-7 2014 Although partial inhibition of human hepatic and intestinal microsomal CYP2C8, CYP2B6, CYP3A4, CYP2D6 and CYP2C19 by GTE catechins was observed in vitro, a clinical study of drug bioavailability attributed a small risk of increased plasma drug levels only for substrates metabolized by CYP3A4, lacking clinical relevance. Catechin 121-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 25456329-8 2014 The presence of the rs3745274 minor allele (CYP2B6 515G>T) reduced CL/F by up to 20% for S-methadone only. Methadone 92-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 25249693-8 2014 UTL-5g and DCA competitively inhibited microsomal CYP1A2, CYP2B6, and CYP2C19 (IC50 values <50 microM), and exhibited time-dependent inhibition of microsomal CYP1A2 with the inactivation efficiency (kinact/KI) of 0.68 and 0.51 minute(-1) mM(-1), respectively. 2,4-dichloroaniline 11-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 25394049-8 2014 Whilst CSF efavirenz concentration was significantly associated with plasma concentration (P<0.001) and CYP2B6 genotype (CSF efavirenz GG to GT/TT GM ratio 0.56, 90% CI 0.42-0.74), CSF 8OH-efavirenz concentration was not (P=0.242 for association with plasma concentration and CSF 8OH-efavirenz GG to GT/TT GM ratio 1.52, 90% CI 0.97-2.36). efavirenz 11-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 25293588-0 2014 Lipoxygenase-catalyzed transformation of epoxy fatty acids to hydroxy-endoperoxides: a potential P450 and lipoxygenase interaction. epoxy fatty acids 41-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-118 25293588-0 2014 Lipoxygenase-catalyzed transformation of epoxy fatty acids to hydroxy-endoperoxides: a potential P450 and lipoxygenase interaction. hydroxy-endoperoxides 62-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-118 25427198-5 2014 The Km and Vmax values of MH were 10.3+-1.3 microM and 99.1+-3.3 nmol/mg protein/min, respectively, for the HLMs; 8.0+-1.6 microM and 112.4+-5.7 nmol/nmol P450/min, respectively, for CYP1A2; and 25.9+-6.6 microM and 134.3+-12.4 nmol/nmol P450/min, respectively, for CYP2C19. meranzin hydrate 26-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-163 25427198-5 2014 The Km and Vmax values of MH were 10.3+-1.3 microM and 99.1+-3.3 nmol/mg protein/min, respectively, for the HLMs; 8.0+-1.6 microM and 112.4+-5.7 nmol/nmol P450/min, respectively, for CYP1A2; and 25.9+-6.6 microM and 134.3+-12.4 nmol/nmol P450/min, respectively, for CYP2C19. meranzin hydrate 26-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 238-246 25395831-3 2014 Honokiol treatment at the highest concentration tested (50 muM) increased the CYP2B6 mRNA level and CYP2B6-catalyzed bupropion hydroxylase activity more than two-fold in three different hepatocyte cultures, indicating that honokiol induces CYP2B6 at higher concentrations. honokiol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 25395831-3 2014 Honokiol treatment at the highest concentration tested (50 muM) increased the CYP2B6 mRNA level and CYP2B6-catalyzed bupropion hydroxylase activity more than two-fold in three different hepatocyte cultures, indicating that honokiol induces CYP2B6 at higher concentrations. honokiol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 25395831-3 2014 Honokiol treatment at the highest concentration tested (50 muM) increased the CYP2B6 mRNA level and CYP2B6-catalyzed bupropion hydroxylase activity more than two-fold in three different hepatocyte cultures, indicating that honokiol induces CYP2B6 at higher concentrations. honokiol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 25395831-6 2014 These results indicate that honokiol is a weak CYP2B6 inducer and is unlikely to increase the metabolism of concomitant CYP2B6 substrates and cause pharmacokinetic-based drug interactions in humans. honokiol 28-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 25096076-7 2014 Efavirenz treatment in arm 1 resulted in 1.74 (relative standard error = 15%), 3.3 (relative standard error = 33.1%) and 4.0 (relative standard error = 37.1%) average fold induction of CYP3A for extensive (CYP2B6*1/*1), intermediate (CYP2B6*1/*6) and slow (CYP2B6*6/*6) efavirenz metabolizers, respectively. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 206-212 25096076-7 2014 Efavirenz treatment in arm 1 resulted in 1.74 (relative standard error = 15%), 3.3 (relative standard error = 33.1%) and 4.0 (relative standard error = 37.1%) average fold induction of CYP3A for extensive (CYP2B6*1/*1), intermediate (CYP2B6*1/*6) and slow (CYP2B6*6/*6) efavirenz metabolizers, respectively. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 234-240 25096076-7 2014 Efavirenz treatment in arm 1 resulted in 1.74 (relative standard error = 15%), 3.3 (relative standard error = 33.1%) and 4.0 (relative standard error = 37.1%) average fold induction of CYP3A for extensive (CYP2B6*1/*1), intermediate (CYP2B6*1/*6) and slow (CYP2B6*6/*6) efavirenz metabolizers, respectively. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 234-240 25096076-10 2014 CONCLUSIONS: Our results indicate that efavirenz induction of CYP3A is influenced by CYP2B6 genetic polymorphisms and that efavirenz/rifampicin co-treatment results in higher induction than efavirenz alone. efavirenz 39-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 24891466-2 2014 Results provide evidence for the possible link between CYP2A6 and CYP2B6 polymorphisms and plasma concentrations of artesunate/dihydroartemisinin and treatment response. artesunate/dihydroartemisinin 116-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 25458686-2 2014 The stereo specific metabolic activity of human CYP-2B6 (cytochrome P450) on endosulfan has been well demonstrated. Endosulfan 77-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-55 25458686-4 2014 The functional similarity was studied at organism level by batch-scale studies and it was proved that B. megaterium could metabolize endosulfan to endosulfan sulfate, as CYP-2B6 does in human system. Endosulfan 133-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-177 24548191-6 2014 Fenofibrate was a potent in vitro inhibitor of CYP2C19 (IC50 = 0.2 mum) and CYP2B6 (IC50 = 0.7 mum) and a moderate inhibitor of CYP2C9 (IC50 = 9.7 mum). Fenofibrate 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 24893120-3 2014 This method does not require any protein-tagging system for protein isolation and has a further advantage that the purification is concomitantly conducted with reconstitution of the enzymes into a phospholipid environment, which is crucial for the catalytic activity assay of P450 enzyme. Phospholipids 197-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 276-280 25076120-3 2014 Emerging evidence suggests that sex steroid bioavailability in the endometrium is determined by adjusting their metabolic rate and fate via regulation of cytochrome (CYP) p450 enzymes. Steroids 36-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-175 25309681-0 2014 Structure-Activity Studies Reveal the Oxazinone Ring Is a Determinant of Cytochrome P450 2B6 Activity Toward Efavirenz. oxazinone 38-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-92 25309681-1 2014 Cytochrome P450 2B6 (CYP2B6) is primarily responsible for the metabolism of the anti-HIV drug efavirenz (EFV). efavirenz 105-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 25309681-1 2014 Cytochrome P450 2B6 (CYP2B6) is primarily responsible for the metabolism of the anti-HIV drug efavirenz (EFV). efavirenz 105-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 25309681-2 2014 We set out to explore the molecular basis for CYP2B6 activity toward EFV by examining the metabolism of eight EFV analogues. efavirenz 69-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 25309681-3 2014 cDNA-expressed CYP2B6 formed monooxygenated metabolites from EFV analogues containing an intact oxazinone or oxazine ring, but not from analogues with a disrupted ring, suggesting this ring is important for metabolism of EFV by CYP2B6. oxazinone 96-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 25309681-3 2014 cDNA-expressed CYP2B6 formed monooxygenated metabolites from EFV analogues containing an intact oxazinone or oxazine ring, but not from analogues with a disrupted ring, suggesting this ring is important for metabolism of EFV by CYP2B6. Oxazines 109-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 25309681-4 2014 Subsequent substrate depletion analysis of EFV and EFV analogues found to be CYP2B6 substrates revealed further differences between these CYP2B6 substrates. efavirenz 43-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 25309681-4 2014 Subsequent substrate depletion analysis of EFV and EFV analogues found to be CYP2B6 substrates revealed further differences between these CYP2B6 substrates. efavirenz 43-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 25309681-4 2014 Subsequent substrate depletion analysis of EFV and EFV analogues found to be CYP2B6 substrates revealed further differences between these CYP2B6 substrates. efavirenz 51-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 25309681-4 2014 Subsequent substrate depletion analysis of EFV and EFV analogues found to be CYP2B6 substrates revealed further differences between these CYP2B6 substrates. efavirenz 51-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 25309681-5 2014 Compounds that were not found to be CYP2B6 substrates were still able to inhibit CYP2B6 activity toward a known substrate, bupropion, suggesting they do gain access to the CYP2B6 active site. Bupropion 123-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 25309681-5 2014 Compounds that were not found to be CYP2B6 substrates were still able to inhibit CYP2B6 activity toward a known substrate, bupropion, suggesting they do gain access to the CYP2B6 active site. Bupropion 123-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 25309681-5 2014 Compounds that were not found to be CYP2B6 substrates were still able to inhibit CYP2B6 activity toward a known substrate, bupropion, suggesting they do gain access to the CYP2B6 active site. Bupropion 123-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 25309681-6 2014 Taken together, these data reveal structural characteristics of EFV, namely, the oxazinone ring, that are critical for CYP2B6 metabolism of compounds with the EFV chemical scaffold. oxazinone 81-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 25017465-1 2014 Cytochrome P450 (P450 or CYP) 46A1 is expressed in brain and has been characterized by its ability to oxidize cholesterol to 24S-hydroxycholesterol. Cholesterol 110-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-34 25017465-1 2014 Cytochrome P450 (P450 or CYP) 46A1 is expressed in brain and has been characterized by its ability to oxidize cholesterol to 24S-hydroxycholesterol. 24-hydroxycholesterol 125-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-34 24664872-13 2014 Further assessment of the mechanism of the effect of alcohol use on adverse experiences, including analysis of CYP2B6 genotype and plasma efavirenz concentrations, is warranted. Alcohols 53-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 24820076-0 2014 Isoniazid mediates the CYP2B6*6 genotype-dependent interaction between efavirenz and antituberculosis drug therapy through mechanism-based inactivation of CYP2A6. Isoniazid 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 24820076-0 2014 Isoniazid mediates the CYP2B6*6 genotype-dependent interaction between efavirenz and antituberculosis drug therapy through mechanism-based inactivation of CYP2A6. efavirenz 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 24820076-4 2014 In vitro studies were conducted to determine whether one or more of the antituberculosis drugs (rifampin, isoniazid, pyrazinamide, or ethambutol) potently inhibit efavirenz 8-hydroxylation by CYP2B6 or efavirenz 7-hydroxylation by CYP2A6, the main mechanisms of efavirenz clearance. Rifampin 96-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 24820076-4 2014 In vitro studies were conducted to determine whether one or more of the antituberculosis drugs (rifampin, isoniazid, pyrazinamide, or ethambutol) potently inhibit efavirenz 8-hydroxylation by CYP2B6 or efavirenz 7-hydroxylation by CYP2A6, the main mechanisms of efavirenz clearance. Ethambutol 134-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 24748562-6 2014 Desmethyl-dabrafenib inhibited CYP2B6, 2C8, 2C9, 2C19, and 3A4 (midazolam, atorvastatin, and nifedipine) with calculated IC50 values of 78, 47, 6.3, 36, 17, 20, and 28 muM, respectively. Desmethyl dabrafenib 0-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 24748562-7 2014 At 30 muM dabrafenib showed increases in CYP2B6 and CYP3A4 mRNA expression indicative of induction. dabrafenib 10-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 24832206-0 2014 Interactions of endosulfan and methoxychlor involving CYP3A4 and CYP2B6 in human HepaRG cells. Endosulfan 16-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 24832206-0 2014 Interactions of endosulfan and methoxychlor involving CYP3A4 and CYP2B6 in human HepaRG cells. Methoxychlor 31-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 24832206-3 2014 In the present work, we searched for interactions between endosulfan and methoxychlor, two organochlorine pesticides whose major routes of metabolism involve CAR- and PXR-regulated CYP3A4 and CYP2B6, and whose mechanisms of action in humans remain poorly understood. Endosulfan 58-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 24832206-3 2014 In the present work, we searched for interactions between endosulfan and methoxychlor, two organochlorine pesticides whose major routes of metabolism involve CAR- and PXR-regulated CYP3A4 and CYP2B6, and whose mechanisms of action in humans remain poorly understood. Methoxychlor 73-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 24832206-3 2014 In the present work, we searched for interactions between endosulfan and methoxychlor, two organochlorine pesticides whose major routes of metabolism involve CAR- and PXR-regulated CYP3A4 and CYP2B6, and whose mechanisms of action in humans remain poorly understood. Hydrocarbons, Chlorinated 91-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 24832206-10 2014 Despite CYP2B6 activity being unchanged and increased with endosulfan and methoxychlor, respectively, no change was observed with their mixture, supporting an antagonistic effect. Methoxychlor 74-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 24452068-13 2014 Potential sex differences in bupropion pharmacokinetics, probably due to differential activities of CYP2B6, should be taken into account when the drug is prescribed. Bupropion 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 24812009-9 2014 Additionally, Aroclor 1260 induced CYP2B6 in primary hepatocytes. aroclor 1260 14-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 24768782-1 2014 BACKGROUND: Cyclophosphamide (CP), an alkylating chemotherapeutic drug, is catalyzed by the Phase I cytochrome P450 (CYPs) isozymes - CYP3A4, CYP3A5, CYP2B6, CYP2C8, CYP2C9 and CYP2C19. Cyclophosphamide 12-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 24658455-1 2014 The glycogen synthase kinase-3 inhibitor LY2090314 specifically impaired CYP2B6 activity during in vitro evaluation of cytochrome P450 (P450) enzyme induction in human hepatocytes. 3-(9-fluoro-2-(piperidin-1-ylcarbonyl)-1,2,3,4-tetrahydro(1,4)diazepino(6,7,1-hi)indol-7-yl)-4-imidazo(1,2-a)pyridin-3-yl-1H-pyrrole-2,5-dione 41-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 24889062-3 2014 The effect of QHS-PQ on the activity of the CYP2B6 and CYP3A4 was also investigated. qhs-pq 14-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 24889062-10 2014 The enzyme activity of CYP2B6 and CYP3A4 increased 2.1-fold and 3.2-fold, respectively, after two-day oral doses of QHS-PQ. qhs-pq 116-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 24889062-13 2014 The enzyme activity of CYP2B6 and CYP3A4 was induced after the two-day oral doses of QHS-PQ. qhs-pq 85-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 24762860-0 2014 Cytochrome p450 gene variants, race, and mortality among clopidogrel-treated patients after acute myocardial infarction. Clopidogrel 57-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 24658455-2 2014 CYP2B6 catalytic activity was significantly decreased following 3-day incubation with 0.1-10 muM LY2090314, on average by 64.3% +- 5.0% at 10 muM. 3-(9-fluoro-2-(piperidin-1-ylcarbonyl)-1,2,3,4-tetrahydro(1,4)diazepino(6,7,1-hi)indol-7-yl)-4-imidazo(1,2-a)pyridin-3-yl-1H-pyrrole-2,5-dione 97-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 24658455-6 2014 Instead, LY2090314 significantly reduced CYP2B6 mRNA levels (Imax = 61.9% +- 1.4%; IC50 = 0.049 +- 0.043 muM), which were significantly correlated with catalytic activity (r(2) = 0.87, slope = 0.77; Imax = 57.0% +- 10.8%, IC50 = 0.057 +- 0.027 muM). 3-(9-fluoro-2-(piperidin-1-ylcarbonyl)-1,2,3,4-tetrahydro(1,4)diazepino(6,7,1-hi)indol-7-yl)-4-imidazo(1,2-a)pyridin-3-yl-1H-pyrrole-2,5-dione 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 24658455-9 2014 CYP2B6 suppression by LY2090314 is not expected clinically, with a projected <1% decrease in hepatic enzyme activity and <1% decrease in hydroxybupropion exposure following bupropion coadministration. 3-(9-fluoro-2-(piperidin-1-ylcarbonyl)-1,2,3,4-tetrahydro(1,4)diazepino(6,7,1-hi)indol-7-yl)-4-imidazo(1,2-a)pyridin-3-yl-1H-pyrrole-2,5-dione 22-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 24658455-9 2014 CYP2B6 suppression by LY2090314 is not expected clinically, with a projected <1% decrease in hepatic enzyme activity and <1% decrease in hydroxybupropion exposure following bupropion coadministration. hydroxybupropion 143-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 24658455-9 2014 CYP2B6 suppression by LY2090314 is not expected clinically, with a projected <1% decrease in hepatic enzyme activity and <1% decrease in hydroxybupropion exposure following bupropion coadministration. Bupropion 150-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 24658455-10 2014 However, simulations showed that observed CYP2B6 suppression could be clinically relevant for a drug with different pharmacokinetic properties from LY2090314. 3-(9-fluoro-2-(piperidin-1-ylcarbonyl)-1,2,3,4-tetrahydro(1,4)diazepino(6,7,1-hi)indol-7-yl)-4-imidazo(1,2-a)pyridin-3-yl-1H-pyrrole-2,5-dione 148-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 24857912-3 2014 A set of genetic variants known for causing variations in clopidogrel responses was selected, which included CYP2C19*2, *3, *17, CYP2B6*4, *6, *9, CYP3A4*18, CYP3A5*3, MDR1 2677G>T/A, 3435C>T, and P2Y12 H2 (742T>C). Clopidogrel 58-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 24500242-2 2014 By contrast with the general reductive oxygen activation pathway of P450s, an H2O2-shunt pathway does not require any supply of electrons and protons for the generation of a highly reactive intermediate (compound I). Oxygen 39-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-73 24500242-2 2014 By contrast with the general reductive oxygen activation pathway of P450s, an H2O2-shunt pathway does not require any supply of electrons and protons for the generation of a highly reactive intermediate (compound I). Hydrogen Peroxide 78-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-73 24500242-4 2014 This review focuses on the P450-catalyzed monooxygenation of organic molecules using H2O2 as the oxidant. Hydrogen Peroxide 85-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 24796891-3 2014 Using computational docking, we predicted the structure of the complex between the antimalarial agent artemether and CYP2B6 whose conformations were obtained by MD simulation. Artemether 102-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 24789201-7 2014 MG132 treatment of HepG2 also attenuated of TCPOBOP-induced CAR transcriptional activation on reporter constructs which contain CAR-binding DNA elements derived from the human CYP2B6 gene. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-5 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 176-182 24738993-4 2014 The enzyme aromatase P450 is expressed aberrantly in endometriosis and is stimulated by prostaglandin E2 , resulting in production of estrogen that induces prostaglandin E2 expression within endometriotic lesions. Dinoprostone 88-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 24885815-0 2014 Genetic variation in the CYP2B6 gene is related to circulating 2,2",4,4"-tetrabromodiphenyl ether (BDE-47) concentrations: an observational population-based study. 2,2',4,4'-tetrabromodiphenyl ether 63-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 24885815-1 2014 BACKGROUND: Since human CYP2B6 has been identified as the major CYP enzyme involved in the metabolism of 2,2",4,4"-tetrabromodiphenyl ether (BDE-47) and that human 2B6 is a highly polymorphic CYP, with known functional variants, we evaluated if circulating concentrations of a major brominated flame retardant, BDE-47, were related to genetic variation in the CYP2B6 gene in a population sample. 2,2',4,4'-tetrabromodiphenyl ether 105-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 24885815-1 2014 BACKGROUND: Since human CYP2B6 has been identified as the major CYP enzyme involved in the metabolism of 2,2",4,4"-tetrabromodiphenyl ether (BDE-47) and that human 2B6 is a highly polymorphic CYP, with known functional variants, we evaluated if circulating concentrations of a major brominated flame retardant, BDE-47, were related to genetic variation in the CYP2B6 gene in a population sample. 2,2',4,4'-tetrabromodiphenyl ether 105-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 360-366 24885815-4 2014 RESULTS: Several SNPs in the CYP2B6 gene were associated with circulating concentrations of BDE-47 (P = 10-4 to 10-9). 2,2',4,4'-tetrabromodiphenyl ether 92-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 24738993-4 2014 The enzyme aromatase P450 is expressed aberrantly in endometriosis and is stimulated by prostaglandin E2 , resulting in production of estrogen that induces prostaglandin E2 expression within endometriotic lesions. Dinoprostone 156-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 24598282-9 2014 In vitro phenotyping studies demonstrated that multiple cytochrome P450 (P450) isoforms, mainly CYP3A4/5 and CYP1A2, were involved in the metabolism of allitinib. AST 1306 152-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-71 24598282-9 2014 In vitro phenotyping studies demonstrated that multiple cytochrome P450 (P450) isoforms, mainly CYP3A4/5 and CYP1A2, were involved in the metabolism of allitinib. AST 1306 152-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 24553381-5 2014 The results revealed that CYP1A2, CYP2B6, and CYP3A4 were induced (>2.0-fold) by omeprazole, phenobarbital, and rifampicin, respectively, in all the hepatocyte lots tested at enzyme activity level (23 lots) and mRNA level (8 lots). Omeprazole 84-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 24598282-11 2014 The formation of a glutathione conjugate of allitinib was independent of NADPH and P450 isoforms, but was catalyzed by glutathione-S-transferase. Glutathione 19-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-87 24553381-5 2014 The results revealed that CYP1A2, CYP2B6, and CYP3A4 were induced (>2.0-fold) by omeprazole, phenobarbital, and rifampicin, respectively, in all the hepatocyte lots tested at enzyme activity level (23 lots) and mRNA level (8 lots). Phenobarbital 96-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 24553381-5 2014 The results revealed that CYP1A2, CYP2B6, and CYP3A4 were induced (>2.0-fold) by omeprazole, phenobarbital, and rifampicin, respectively, in all the hepatocyte lots tested at enzyme activity level (23 lots) and mRNA level (8 lots). Rifampin 115-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 24598282-11 2014 The formation of a glutathione conjugate of allitinib was independent of NADPH and P450 isoforms, but was catalyzed by glutathione-S-transferase. AST 1306 44-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-87 24390631-6 2014 Ethnic differences and the associated prevalence of CYP2B6 polymorphisms result in significant differences in the PKPD associated with a standard 600 mg per day dose of EFV, warranting dosage reduction in carriers of specific CYP2B6 polymorphisms. efavirenz 169-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 24956253-2 2014 The aim of this study was to evaluate the clinical and economic impact of efavirenz (EFV) dose adjustment by monitoring plasma concentrations and pharmacogenetic analysis of the 516G>T CYP2B6 polymorphism. efavirenz 74-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 188-194 24956253-2 2014 The aim of this study was to evaluate the clinical and economic impact of efavirenz (EFV) dose adjustment by monitoring plasma concentrations and pharmacogenetic analysis of the 516G>T CYP2B6 polymorphism. efavirenz 85-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 188-194 24548980-12 2014 The relationships between transcript expression and patient-level data demonstrated; ABCC2 expression was different by race (1.26 +- 1.82 Caucasian versus 1.37 +- 0.86 non-Caucasian; p = 0.049) and CYP2B6 expression was different by treatment (2.07 +- 2.94 cyclophosphamide versus 0.45 +- 0.5 mycophenolate; p = 0.01). Cyclophosphamide 257-273 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 24548980-12 2014 The relationships between transcript expression and patient-level data demonstrated; ABCC2 expression was different by race (1.26 +- 1.82 Caucasian versus 1.37 +- 0.86 non-Caucasian; p = 0.049) and CYP2B6 expression was different by treatment (2.07 +- 2.94 cyclophosphamide versus 0.45 +- 0.5 mycophenolate; p = 0.01). Mycophenolic Acid 293-306 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 24390631-6 2014 Ethnic differences and the associated prevalence of CYP2B6 polymorphisms result in significant differences in the PKPD associated with a standard 600 mg per day dose of EFV, warranting dosage reduction in carriers of specific CYP2B6 polymorphisms. efavirenz 169-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 226-232 24445070-0 2014 The role of cytochrome P450 2B6 and 2B4 substrate access channel residues predicted based on crystal structures of the amlodipine complexes. Amlodipine 119-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-39 23629159-2 2014 We hypothesized that CYP2C19 and CYP2B6 genetic polymorphisms influence the extent of induction of omeprazole metabolism by efavirenz. Omeprazole 99-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 23629159-2 2014 We hypothesized that CYP2C19 and CYP2B6 genetic polymorphisms influence the extent of induction of omeprazole metabolism by efavirenz. efavirenz 124-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 24568283-0 2014 Generation of complexity in fungal terpene biosynthesis: discovery of a multifunctional cytochrome P450 in the fumagillin pathway. Terpenes 35-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-103 24568283-5 2014 Most significantly, the P450 monooxygenase is shown to catalyze successive hydroxylation, bicyclic ring-opening, and two epoxidations that generate the sesquiterpenoid core skeleton of 1. sesquiterpenoid 152-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 24554642-4 2014 The engineered P450 is the first enzyme for this type of highly selective alkane hydroxylation, being useful for the C-H activation and functionalization of alkanes and the preparation of enantiopure alcohols. Alkanes 74-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 24554642-4 2014 The engineered P450 is the first enzyme for this type of highly selective alkane hydroxylation, being useful for the C-H activation and functionalization of alkanes and the preparation of enantiopure alcohols. Alkanes 157-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 24554642-4 2014 The engineered P450 is the first enzyme for this type of highly selective alkane hydroxylation, being useful for the C-H activation and functionalization of alkanes and the preparation of enantiopure alcohols. Alcohols 200-208 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 23834474-5 2014 For verification, the pregnancy-related changes in the disposition of methadone (cleared by CYP2B6, 3A and 2C19) and glyburide (cleared by CYP3A, 2C9 and 2C19) were predicted. Methadone 70-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 24445070-1 2014 Recent X-ray crystal structures of human cytochrome P450 2B6 and rabbit cytochrome P450 2B4 in complex with amlodipine showed two bound ligand molecules, one in the active site and one in the substrate access channel. Amlodipine 108-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-60 24316028-3 2014 We investigated in patients co-infected with human immunodeficiency virus (HIV) and TB recruited in Rwanda the effects of 10 SNPs in five drug-metabolizing enzymes on EFV plasma levels and treatment response when patients are treated with EFV-containing therapy alone (n=28) and when combined with rifampicin-based TB treatment (n=62), and the validity of genotyping for CYP2B6 single nucleotide polymorphisms in predicting supra-therapeutic EFV levels. efavirenz 239-242 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 371-377 24359841-13 2014 By multivariate analysis, factors associated with high ALP, total bilirubin and direct bilirubin included CYP2B6 haplotype *6/*6, high serum ALP at Week 0 and positive anti-HCV (all P<0.05). Bilirubin 87-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 24316028-3 2014 We investigated in patients co-infected with human immunodeficiency virus (HIV) and TB recruited in Rwanda the effects of 10 SNPs in five drug-metabolizing enzymes on EFV plasma levels and treatment response when patients are treated with EFV-containing therapy alone (n=28) and when combined with rifampicin-based TB treatment (n=62), and the validity of genotyping for CYP2B6 single nucleotide polymorphisms in predicting supra-therapeutic EFV levels. efavirenz 239-242 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 371-377 24316028-6 2014 Predictive factors of EFV plasma levels in the presence of rifampicin-based TB treatment were CYP2A6 1093G>A, CYP2B6 516G>T, and CYP2B6 983T>C accounting for 27%, 43%, and 29% of the total variance in EFV levels, respectively. efavirenz 22-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 24316028-7 2014 There was a high positive predictive value (PPV) (100%) for CYP2B6 516T/T and 983T/T genotypes in predicting EFV plasma levels above the therapeutic range, but this PPV decreased in the presence of rifampicin-based TB treatment. efavirenz 109-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 24316028-5 2014 CYP2B6 516T/T genotype was associated with high EFV levels compared to other CYP2B6 516G>T genotypes in the presence and in the absence of rifampicin-based TB treatment. Rifampin 142-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 24316028-6 2014 Predictive factors of EFV plasma levels in the presence of rifampicin-based TB treatment were CYP2A6 1093G>A, CYP2B6 516G>T, and CYP2B6 983T>C accounting for 27%, 43%, and 29% of the total variance in EFV levels, respectively. efavirenz 22-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 24186263-10 2014 RESULTS: In vitro AZD2066 inhibited CYP1A2, CYP2B6, CYP2C9, CYP2C19 and CYP2D6. AZD2066 18-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 24497997-8 2014 RESULTS: EFV apparent clearance (CL/F) was 2.2 and 1.74 fold higher in CYP2B6*6 (*1/*1) and CYP2B6*6 (*1/*6) compared CYP2B6*6 (*6/*6) carriers, while a 22% increase in F1 was observed for carriers of ABCB1 c.4046A>G variant allele. efavirenz 9-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 24252946-0 2014 Metformin represses drug-induced expression of CYP2B6 by modulating the constitutive androstane receptor signaling. Metformin 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 24252946-4 2014 We show that metformin could suppress drug-induced expression of CYP2B6 (a typical target gene of CAR) by modulating the phosphorylation status of CAR. Metformin 13-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 24252946-5 2014 In human hepatocytes, metformin robustly suppressed the expression of CYP2B6 induced by both indirect (phenobarbital) and direct CITCO [6-(4-chlorophenyl)imidazo[2,1-b]1,3thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime] activators of human CAR. Metformin 22-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 24252946-5 2014 In human hepatocytes, metformin robustly suppressed the expression of CYP2B6 induced by both indirect (phenobarbital) and direct CITCO [6-(4-chlorophenyl)imidazo[2,1-b]1,3thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime] activators of human CAR. Phenobarbital 103-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 24252946-5 2014 In human hepatocytes, metformin robustly suppressed the expression of CYP2B6 induced by both indirect (phenobarbital) and direct CITCO [6-(4-chlorophenyl)imidazo[2,1-b]1,3thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime] activators of human CAR. citco [6-(4-chlorophenyl)imidazo[2,1-b]1,3thiazole-5-carbaldehyde o-(3,4-dichlorobenzyl)oxime 129-222 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 24252946-9 2014 Our results suggest that metformin is a potent repressor of drug-induced CYP2B6 expression through specific inhibition of human CAR activation. Metformin 25-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 24252946-10 2014 Thus, metformin may affect the metabolism and clearance of drugs that are CYP2B6 substrates. Metformin 6-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 23990572-0 2014 Dependence of efavirenz- and rifampicin-isoniazid-based antituberculosis treatment drug-drug interaction on CYP2B6 and NAT2 genetic polymorphisms: ANRS 12154 study in Cambodia. efavirenz 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 23990572-0 2014 Dependence of efavirenz- and rifampicin-isoniazid-based antituberculosis treatment drug-drug interaction on CYP2B6 and NAT2 genetic polymorphisms: ANRS 12154 study in Cambodia. Rifampin 29-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 23990572-0 2014 Dependence of efavirenz- and rifampicin-isoniazid-based antituberculosis treatment drug-drug interaction on CYP2B6 and NAT2 genetic polymorphisms: ANRS 12154 study in Cambodia. Isoniazid 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 24497997-8 2014 RESULTS: EFV apparent clearance (CL/F) was 2.2 and 1.74 fold higher in CYP2B6*6 (*1/*1) and CYP2B6*6 (*1/*6) compared CYP2B6*6 (*6/*6) carriers, while a 22% increase in F1 was observed for carriers of ABCB1 c.4046A>G variant allele. efavirenz 9-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 24497997-8 2014 RESULTS: EFV apparent clearance (CL/F) was 2.2 and 1.74 fold higher in CYP2B6*6 (*1/*1) and CYP2B6*6 (*1/*6) compared CYP2B6*6 (*6/*6) carriers, while a 22% increase in F1 was observed for carriers of ABCB1 c.4046A>G variant allele. efavirenz 9-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 24497997-8 2014 RESULTS: EFV apparent clearance (CL/F) was 2.2 and 1.74 fold higher in CYP2B6*6 (*1/*1) and CYP2B6*6 (*1/*6) compared CYP2B6*6 (*6/*6) carriers, while a 22% increase in F1 was observed for carriers of ABCB1 c.4046A>G variant allele. Fluorine 10-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 24497997-8 2014 RESULTS: EFV apparent clearance (CL/F) was 2.2 and 1.74 fold higher in CYP2B6*6 (*1/*1) and CYP2B6*6 (*1/*6) compared CYP2B6*6 (*6/*6) carriers, while a 22% increase in F1 was observed for carriers of ABCB1 c.4046A>G variant allele. Fluorine 10-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 24497997-8 2014 RESULTS: EFV apparent clearance (CL/F) was 2.2 and 1.74 fold higher in CYP2B6*6 (*1/*1) and CYP2B6*6 (*1/*6) compared CYP2B6*6 (*6/*6) carriers, while a 22% increase in F1 was observed for carriers of ABCB1 c.4046A>G variant allele. Fluorine 10-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 24262967-0 2014 Effects of CYP3A5, CYP2C19, and CYP2B6 on the clinical efficacy and adverse outcomes of sibutramine therapy: a crucial role for the CYP2B6*6 allele. sibutramine 88-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 24489693-0 2014 Impact of ABCB1 and CYP2B6 genetic polymorphisms on methadone metabolism, dose and treatment response in patients with opioid addiction: a systematic review and meta-analysis. Methadone 52-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 24489693-2 2014 OBJECTIVES: To determine whether the CYP2B6*6 or ABCB1 (rs1045642) polymorphisms are associated with variation in methadone response (plasma concentration, dose, or response to treatment). Methadone 114-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 24489693-4 2014 We included studies that reported methadone plasma concentration, methadone response, or methadone dose in relation to the CYP2B6*6 or ABCB1 polymorphisms. Methadone 34-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-129 24489693-7 2014 Trough (R) methadone plasma concentration was significantly higher in CYP2B6*6 homozygous carriers when compared to non-carriers (standardized mean difference [SMD] = 0.53, 95% confidence interval [CI], 0.05-1.00, p = 0.03) with minimal heterogeneity (I(2) = 0%). Methadone 11-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 24489693-11 2014 CONCLUSION: Although the number of studies included and sample size were modest, this is the first meta analysis to show participants homozygous for the CYP2B6*6 genotype have higher trough (R) and (S) methadone plasma concentrations, suggesting that methadone metabolism is significantly slower in *6 homozygous carriers. Methadone 202-211 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 24489693-11 2014 CONCLUSION: Although the number of studies included and sample size were modest, this is the first meta analysis to show participants homozygous for the CYP2B6*6 genotype have higher trough (R) and (S) methadone plasma concentrations, suggesting that methadone metabolism is significantly slower in *6 homozygous carriers. Methadone 251-260 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 24387788-0 2014 Effects of anthocyanidins and anthocyanins on the expression and catalytic activities of CYP2A6, CYP2B6, CYP2C9, and CYP3A4 in primary human hepatocytes and human liver microsomes. Anthocyanins 11-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 24387788-0 2014 Effects of anthocyanidins and anthocyanins on the expression and catalytic activities of CYP2A6, CYP2B6, CYP2C9, and CYP3A4 in primary human hepatocytes and human liver microsomes. Anthocyanins 30-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 24387788-3 2014 The current study evaluated the effects of anthocyanidins and anthocyanins on the expression and catalytic activity of major drug-metabolizing enzymes CYP2C9, CYP2A6, CYP2B6, and CYP3A4 in primary cultures of human hepatocytes and human liver microsomes. Anthocyanins 43-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-173 24387788-3 2014 The current study evaluated the effects of anthocyanidins and anthocyanins on the expression and catalytic activity of major drug-metabolizing enzymes CYP2C9, CYP2A6, CYP2B6, and CYP3A4 in primary cultures of human hepatocytes and human liver microsomes. Anthocyanins 62-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-173 24387788-8 2014 Catalytic activities of CYP2C9, CYP2A6, CYP2B6, and CYP3A4 enzymes were inhibited by all anthocyanidins to different extents (e.g., delphinidin inhibits CYP3A4 by >90% at 100 muM with IC50 = 32 muM). Anthocyanins 89-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 24387788-8 2014 Catalytic activities of CYP2C9, CYP2A6, CYP2B6, and CYP3A4 enzymes were inhibited by all anthocyanidins to different extents (e.g., delphinidin inhibits CYP3A4 by >90% at 100 muM with IC50 = 32 muM). delphinidin 132-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 23824676-1 2014 The cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of testosterone. Testosterone 66-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-23 23824676-1 2014 The cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of testosterone. Testosterone 66-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 24262967-0 2014 Effects of CYP3A5, CYP2C19, and CYP2B6 on the clinical efficacy and adverse outcomes of sibutramine therapy: a crucial role for the CYP2B6*6 allele. sibutramine 88-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-138 24262967-9 2014 CONCLUSION: The CYP2B6*6 allele influences the extent of weight reduction and pulse rate changes in patients undergoing sibutramine treatment. sibutramine 120-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 24909419-4 2014 For example, in patients treated with abacavir the screening for HLA-B*5701 before starting treatment is routine clinical practice and standard of care for all patients; efavirenz plasma levels are influenced by single nucleotide polymorphism (SNP) CYP2B6-516G>T (rs3745274). abacavir 38-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 249-255 24423593-2 2014 P-gp (fexofenadine) and CYP-specific substrates (caffeine for CYP1A2, bupropion for CYP2B6, flurbiprofen for CYP2C9, omeprazole for CYP2C19, dextromethorphan for CYP2D6 and midazolam for CYP3A4) and their metabolites were extracted from DBS (10 microl) using methanol. Bupropion 70-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 25366478-5 2014 Delamanid (100 microM) had little potential for mechanism-based inactivation on eight CYP isoforms (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). OPC-67683 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 24153159-0 2014 Association of the CYP2B6 c.516G>T polymorphism with high blood propofol concentrations in women from northern Greece. Propofol 67-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 25429674-2 2014 In in vitro interaction studies, itraconazole, ketoconazole, and miconazole were found to have higher inhibitory effects on cytochrome P450 (P450 or CYP) 3A4 and 3A5 activities than the other azoles or echinocandins did. Itraconazole 33-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-157 25429674-2 2014 In in vitro interaction studies, itraconazole, ketoconazole, and miconazole were found to have higher inhibitory effects on cytochrome P450 (P450 or CYP) 3A4 and 3A5 activities than the other azoles or echinocandins did. Ketoconazole 47-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-157 25429674-2 2014 In in vitro interaction studies, itraconazole, ketoconazole, and miconazole were found to have higher inhibitory effects on cytochrome P450 (P450 or CYP) 3A4 and 3A5 activities than the other azoles or echinocandins did. Miconazole 65-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-157 25429674-2 2014 In in vitro interaction studies, itraconazole, ketoconazole, and miconazole were found to have higher inhibitory effects on cytochrome P450 (P450 or CYP) 3A4 and 3A5 activities than the other azoles or echinocandins did. Echinocandins 202-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-157 24153159-2 2014 In this study we examined the effect of the apparently functional CYP2B6 c.516G>T polymorphism on the distribution of propofol concentrations, quantified by GC/MS analysis following a single bolus dose, in the blood of 44 Greek women undergoing oocyte retrieval. Propofol 121-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 25343299-9 2014 These in vitro results indicate that cedrol, beta-cedrene, and thujopsene need to be examined for potential pharmacokinetic drug interactions in vivo due to their potent inhibition of CYP2B6 and CYP3A4. cedrene 45-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-190 24477223-7 2014 Univariant and multivariant analysis indicated associations for CYP2B6 g.18492T>C (p < 0.001 and p = 0.001), aspartate aminotransferase (AST; p = 0.001 and p = 0.006) and blood urea nitrogen (BUN; p = 0.011 and p = 0.016) with plasma efavirenz concentration. Urea 183-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 24477223-7 2014 Univariant and multivariant analysis indicated associations for CYP2B6 g.18492T>C (p < 0.001 and p = 0.001), aspartate aminotransferase (AST; p = 0.001 and p = 0.006) and blood urea nitrogen (BUN; p = 0.011 and p = 0.016) with plasma efavirenz concentration. Nitrogen 188-196 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 25343291-5 2014 CYP3A4 and CYP3A5 were only involved in the formation of M1, whereas CYP2B6 and CYP2C19 were responsible for all metabolic reactions of sibutramine. sibutramine 136-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 24293093-0 2014 Functional polymorphism of CYP2B6 G15631T is associated with hematologic and cytogenetic response in chronic myeloid leukemia patients treated with imatinib. Imatinib Mesylate 148-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 24293093-3 2014 The present study aimed to understand the functional impact of CYP2B6 15631G>T polymorphism on the response of imatinib in CML patients and its relation to CML susceptibility. Imatinib Mesylate 114-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 24142869-8 2014 The final model included only CYP2B6 genotype as a covariate that predicts EFV clearance through the formation of 8-OH EFV that represented 65% to 80% of EFV clearance. efavirenz 75-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 24142869-8 2014 The final model included only CYP2B6 genotype as a covariate that predicts EFV clearance through the formation of 8-OH EFV that represented 65% to 80% of EFV clearance. 8-hydroxyefavirenz 114-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 24142869-8 2014 The final model included only CYP2B6 genotype as a covariate that predicts EFV clearance through the formation of 8-OH EFV that represented 65% to 80% of EFV clearance. efavirenz 119-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 24142869-9 2014 The total clearance of EFV in CYP2B6*6/*6 genotype was ~30% lower than CYP2B6*1/*1 or CYP2B6*1/*6 alleles (P < .001). efavirenz 23-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 25343299-4 2014 Cedrol, beta-cedrene, and thujopsene were found to be potent competitive inhibitors of CYP2B6-mediated bupropion hydroxylase with inhibition constant (Ki) values of 0.9, 1.6, and 0.8 muM, respectively, comparable with that of a selective CYP2B6 inhibitor, thioTEPA (Ki, 2.9 muM). cedrol 0-6 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 25343299-4 2014 Cedrol, beta-cedrene, and thujopsene were found to be potent competitive inhibitors of CYP2B6-mediated bupropion hydroxylase with inhibition constant (Ki) values of 0.9, 1.6, and 0.8 muM, respectively, comparable with that of a selective CYP2B6 inhibitor, thioTEPA (Ki, 2.9 muM). cedrol 0-6 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 238-244 25343299-4 2014 Cedrol, beta-cedrene, and thujopsene were found to be potent competitive inhibitors of CYP2B6-mediated bupropion hydroxylase with inhibition constant (Ki) values of 0.9, 1.6, and 0.8 muM, respectively, comparable with that of a selective CYP2B6 inhibitor, thioTEPA (Ki, 2.9 muM). cedrene 8-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 25343299-4 2014 Cedrol, beta-cedrene, and thujopsene were found to be potent competitive inhibitors of CYP2B6-mediated bupropion hydroxylase with inhibition constant (Ki) values of 0.9, 1.6, and 0.8 muM, respectively, comparable with that of a selective CYP2B6 inhibitor, thioTEPA (Ki, 2.9 muM). cedrene 8-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 238-244 25343299-4 2014 Cedrol, beta-cedrene, and thujopsene were found to be potent competitive inhibitors of CYP2B6-mediated bupropion hydroxylase with inhibition constant (Ki) values of 0.9, 1.6, and 0.8 muM, respectively, comparable with that of a selective CYP2B6 inhibitor, thioTEPA (Ki, 2.9 muM). thujopsene 26-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 25343299-4 2014 Cedrol, beta-cedrene, and thujopsene were found to be potent competitive inhibitors of CYP2B6-mediated bupropion hydroxylase with inhibition constant (Ki) values of 0.9, 1.6, and 0.8 muM, respectively, comparable with that of a selective CYP2B6 inhibitor, thioTEPA (Ki, 2.9 muM). thujopsene 26-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 238-244 25343299-4 2014 Cedrol, beta-cedrene, and thujopsene were found to be potent competitive inhibitors of CYP2B6-mediated bupropion hydroxylase with inhibition constant (Ki) values of 0.9, 1.6, and 0.8 muM, respectively, comparable with that of a selective CYP2B6 inhibitor, thioTEPA (Ki, 2.9 muM). Thiotepa 256-264 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 25343299-9 2014 These in vitro results indicate that cedrol, beta-cedrene, and thujopsene need to be examined for potential pharmacokinetic drug interactions in vivo due to their potent inhibition of CYP2B6 and CYP3A4. thujopsene 63-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-190 25343299-9 2014 These in vitro results indicate that cedrol, beta-cedrene, and thujopsene need to be examined for potential pharmacokinetic drug interactions in vivo due to their potent inhibition of CYP2B6 and CYP3A4. cedrol 37-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-190 24245489-5 2014 Genotyping for various CYP single-nucleotide polymorphisms showed that the patient carried mutant alleles encoding enzymes CYP2B6 and CYP2C9 involved in prasugrel metabolic pathway. Prasugrel Hydrochloride 153-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-129 24128861-0 2013 CYP2B6*6 is an independent determinant of inferior response to fludarabine plus cyclophosphamide in chronic lymphocytic leukemia. fludarabine 63-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 24128861-0 2013 CYP2B6*6 is an independent determinant of inferior response to fludarabine plus cyclophosphamide in chronic lymphocytic leukemia. Cyclophosphamide 80-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 24128861-2 2013 CYP2B6 is a polymorphic cytochrome P450 isoform that converts cyclophosphamide to its active form. Cyclophosphamide 62-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23860413-3 2013 Microbial fatty acid-hydroxylation enzymes, including P450, lipoxygenase, hydratase, 12-hydroxylase, and diol synthase, synthesize regio-specific hydroxy fatty acids. microbial fatty acid 0-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-72 23860413-3 2013 Microbial fatty acid-hydroxylation enzymes, including P450, lipoxygenase, hydratase, 12-hydroxylase, and diol synthase, synthesize regio-specific hydroxy fatty acids. hydroxy fatty acids 146-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-72 23846872-3 2013 Efavirenz clearance was predictable using in vitro data for carriers of the CYP2B6*1/*1 genotype, but the prediction in carriers of the CYP2B6*6 allele was poor. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 23818090-7 2013 Hydroxy bupropion formation was used as a more sensitive marker of CYP2B6 activity than bupropion kinetics. hydroxybupropion 0-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 23818090-7 2013 Hydroxy bupropion formation was used as a more sensitive marker of CYP2B6 activity than bupropion kinetics. Bupropion 8-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 23057845-3 2013 Our results demonstrate that: (i) the putative hydrophobic residues located in the F-helix and polar residues in I-helix are critical in the herbicide resistance; (ii) the binding mode analysis and binding free energy calculation indicate that the distance between catalytic site of chlortoluron and heme of P450, as well as the binding affinity are key elements affecting the resistance for plants. chlortoluron 283-295 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 308-312 24021950-9 2013 The V(max) of terfenadine hydroxylation in recombinant enzyme was found to be 29.4 pmol/pmol P450 per minute and in the cells 6.0 pmol/pmol P450 per minute. Terfenadine 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 24021950-9 2013 The V(max) of terfenadine hydroxylation in recombinant enzyme was found to be 29.4 pmol/pmol P450 per minute and in the cells 6.0 pmol/pmol P450 per minute. Terfenadine 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-144 23057845-3 2013 Our results demonstrate that: (i) the putative hydrophobic residues located in the F-helix and polar residues in I-helix are critical in the herbicide resistance; (ii) the binding mode analysis and binding free energy calculation indicate that the distance between catalytic site of chlortoluron and heme of P450, as well as the binding affinity are key elements affecting the resistance for plants. Heme 300-304 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 308-312 24260284-0 2013 CYP2B6 non-coding variation associated with smoking cessation is also associated with differences in allelic expression, splicing, and nicotine metabolism independent of common amino-acid changes. Nicotine 135-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 24521642-9 2013 Logistic regression analysis showed CYP2B6 516G>T polymorphism significantly associated with virologic outcome in patients receiving EFV-based regimen. efavirenz 136-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 23089673-6 2013 CYP2B6*6 genotype significantly influenced 4beta-OHC/Chol ratio at weeks 16 (P=0.02) and 48 (P=0.04) being highest in CYP2B6*6/*6>*1/*6>*1/*1. 4beta 43-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23089673-6 2013 CYP2B6*6 genotype significantly influenced 4beta-OHC/Chol ratio at weeks 16 (P=0.02) and 48 (P=0.04) being highest in CYP2B6*6/*6>*1/*6>*1/*1. 4beta 43-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 23089673-6 2013 CYP2B6*6 genotype significantly influenced 4beta-OHC/Chol ratio at weeks 16 (P=0.02) and 48 (P=0.04) being highest in CYP2B6*6/*6>*1/*6>*1/*1. ohc 49-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23089673-6 2013 CYP2B6*6 genotype significantly influenced 4beta-OHC/Chol ratio at weeks 16 (P=0.02) and 48 (P=0.04) being highest in CYP2B6*6/*6>*1/*6>*1/*1. ohc 49-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 23089673-6 2013 CYP2B6*6 genotype significantly influenced 4beta-OHC/Chol ratio at weeks 16 (P=0.02) and 48 (P=0.04) being highest in CYP2B6*6/*6>*1/*6>*1/*1. chol 53-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23089673-6 2013 CYP2B6*6 genotype significantly influenced 4beta-OHC/Chol ratio at weeks 16 (P=0.02) and 48 (P=0.04) being highest in CYP2B6*6/*6>*1/*6>*1/*1. chol 53-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 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 2 subfamily B member 6 Homo sapiens 108-114 25278738-6 2013 We found that the SNPs on CYP2B6 were associated with plasma S-methadone concentration; SNPs on CYP3A4 were associated with withdrawal symptoms and side effects; and SNPs on CYP2C19 were associated with methadone dose. Methadone 63-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 25278738-6 2013 We found that the SNPs on CYP2B6 were associated with plasma S-methadone concentration; SNPs on CYP3A4 were associated with withdrawal symptoms and side effects; and SNPs on CYP2C19 were associated with methadone dose. Methadone 203-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 24260284-1 2013 The Cytochrome P450 2B6 (CYP2B6) enzyme makes a small contribution to hepatic nicotine metabolism relative to CYP2A6, but CYP2B6 is the primary enzyme responsible for metabolism of the smoking cessation drug bupropion. Nicotine 78-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-23 24260284-1 2013 The Cytochrome P450 2B6 (CYP2B6) enzyme makes a small contribution to hepatic nicotine metabolism relative to CYP2A6, but CYP2B6 is the primary enzyme responsible for metabolism of the smoking cessation drug bupropion. Nicotine 78-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 24260284-1 2013 The Cytochrome P450 2B6 (CYP2B6) enzyme makes a small contribution to hepatic nicotine metabolism relative to CYP2A6, but CYP2B6 is the primary enzyme responsible for metabolism of the smoking cessation drug bupropion. Bupropion 208-217 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-23 24260284-1 2013 The Cytochrome P450 2B6 (CYP2B6) enzyme makes a small contribution to hepatic nicotine metabolism relative to CYP2A6, but CYP2B6 is the primary enzyme responsible for metabolism of the smoking cessation drug bupropion. Bupropion 208-217 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 24260284-1 2013 The Cytochrome P450 2B6 (CYP2B6) enzyme makes a small contribution to hepatic nicotine metabolism relative to CYP2A6, but CYP2B6 is the primary enzyme responsible for metabolism of the smoking cessation drug bupropion. Bupropion 208-217 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 24260284-2 2013 Using CYP2A6 genotype as a covariate, we find that a non-coding polymorphism in CYP2B6 previously associated with smoking cessation (rs8109525) is also significantly associated with nicotine metabolism. Nicotine 182-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 23963955-1 2013 Cytochrome P450 (P450) function requires the interaction of P450 and NADPH-cytochrome P450 reductase (CPR) in membranes, and is frequently studied using reconstituted systems composed solely of phosphatidylcholine. Phosphatidylcholines 194-213 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 23963955-1 2013 Cytochrome P450 (P450) function requires the interaction of P450 and NADPH-cytochrome P450 reductase (CPR) in membranes, and is frequently studied using reconstituted systems composed solely of phosphatidylcholine. Phosphatidylcholines 194-213 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 23886934-1 2013 Cytochrome P450 (P450, CYP) 1 family plays a primary role in the detoxification and bioactivation of polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 101-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-29 23982262-10 2013 A trend toward higher nevirapine AUC(0-6 h) for the CYP2B6 516TT (rs3745274; Q172H) genotype was observed in European Americans (P=0.19). Nevirapine 22-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 23774940-0 2013 Association between CYP2B6 polymorphisms and Nevirapine-induced SJS/TEN: a pharmacogenetics study. Nevirapine 45-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 23978092-0 2013 Complexity generation in fungal polyketide biosynthesis: a spirocycle-forming P450 in the concise pathway to the antifungal drug griseofulvin. Polyketides 32-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 23978092-3 2013 In this study, we have systematically characterized each of the biosynthetic enzymes related to the biogenesis of 1, including the characterization of a new polyketide synthase GsfA that synthesizes the benzophenone precursor and a cytochrome P450 GsfF that performs oxidative coupling between the orcinol and the phloroglucinol rings to yield the grisan structure. gsfa 177-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 243-247 23935064-4 2013 Rifampin induced the expression of 10 clinically important and 13 additional P450 genes and repressed the expression of 9 other P450 genes (P < 0.05). Rifampin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-81 23852652-5 2013 Roles for CYP3A4, CYP2C9, CYP2C19 and CYP2B6 in the metabolism of ospemifene and its two main metabolites, 4--hydroxyospemifene and 4"-hydroxyospemifene, were confirmed. Ospemifene 66-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 23852652-5 2013 Roles for CYP3A4, CYP2C9, CYP2C19 and CYP2B6 in the metabolism of ospemifene and its two main metabolites, 4--hydroxyospemifene and 4"-hydroxyospemifene, were confirmed. 4-hydroxyospemifene 107-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Heme 60-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Heme 60-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 150-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 23886699-0 2013 The effect of ritonavir on human CYP2B6 catalytic activity: heme modification contributes to the mechanism-based inactivation of CYP2B6 and CYP3A4 by ritonavir. Ritonavir 150-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 23886699-1 2013 The mechanism-based inactivation of human CYP2B6 by ritonavir (RTV) in a reconstituted system was investigated. Ritonavir 52-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 23886699-1 2013 The mechanism-based inactivation of human CYP2B6 by ritonavir (RTV) in a reconstituted system was investigated. Ritonavir 63-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 23886699-4 2013 Inactivation of CYP2B6 by incubation with 10 muM RTV for 10 min resulted in an approximately 50% loss of catalytic activity and native heme, but no modification of the apoprotein was observed. Ritonavir 49-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 23886699-4 2013 Inactivation of CYP2B6 by incubation with 10 muM RTV for 10 min resulted in an approximately 50% loss of catalytic activity and native heme, but no modification of the apoprotein was observed. Heme 135-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 23935064-4 2013 Rifampin induced the expression of 10 clinically important and 13 additional P450 genes and repressed the expression of 9 other P450 genes (P < 0.05). Rifampin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 23935064-6 2013 Several of these changes were highly negatively correlated with the rifampin-induced changes in the expression of their predicted target P450 mRNAs, supporting the possibility of miRNA-induced regulation of P450 mRNA expression. Rifampin 68-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-141 23935064-6 2013 Several of these changes were highly negatively correlated with the rifampin-induced changes in the expression of their predicted target P450 mRNAs, supporting the possibility of miRNA-induced regulation of P450 mRNA expression. Rifampin 68-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 207-211 23935064-9 2013 In conclusion, rifampin treatment alters miRNA expression patterns in human hepatocytes, and some of the changes were correlated with the rifampin-induced changes in expression of the P450 mRNAs they are predicted to target. Rifampin 15-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-188 23935064-9 2013 In conclusion, rifampin treatment alters miRNA expression patterns in human hepatocytes, and some of the changes were correlated with the rifampin-induced changes in expression of the P450 mRNAs they are predicted to target. Rifampin 138-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-188 24005963-4 2013 Honokiol also moderately inhibited CYP2B6-mediated bupropion hydroxylation and CYP2D6-mediated bufuralol 1"-hydroxylation with K(i) values of 17.5 and 12.0 muM, respectively. honokiol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 23777987-0 2013 Potent inhibition of cytochrome P450 2B6 by sibutramine in human liver microsomes. sibutramine 44-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-40 23777987-3 2013 Sibutramine showed potent inhibition of CYP2B6-mediated bupropion 6-hydroxylation with an IC50 value of 1.61muM and Ki value of 0.466muM in a competitive manner at microsomal protein concentrations of 0.25mg/ml; this was 3.49-fold more potent than the typical CYP2B6 inhibitor thio-TEPA (Ki=1.59muM). sibutramine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 23777987-3 2013 Sibutramine showed potent inhibition of CYP2B6-mediated bupropion 6-hydroxylation with an IC50 value of 1.61muM and Ki value of 0.466muM in a competitive manner at microsomal protein concentrations of 0.25mg/ml; this was 3.49-fold more potent than the typical CYP2B6 inhibitor thio-TEPA (Ki=1.59muM). sibutramine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 260-266 23777987-3 2013 Sibutramine showed potent inhibition of CYP2B6-mediated bupropion 6-hydroxylation with an IC50 value of 1.61muM and Ki value of 0.466muM in a competitive manner at microsomal protein concentrations of 0.25mg/ml; this was 3.49-fold more potent than the typical CYP2B6 inhibitor thio-TEPA (Ki=1.59muM). Bupropion 56-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 23777987-3 2013 Sibutramine showed potent inhibition of CYP2B6-mediated bupropion 6-hydroxylation with an IC50 value of 1.61muM and Ki value of 0.466muM in a competitive manner at microsomal protein concentrations of 0.25mg/ml; this was 3.49-fold more potent than the typical CYP2B6 inhibitor thio-TEPA (Ki=1.59muM). Thiotepa 277-286 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 23777987-5 2013 These observations indicated 35.6- and 33.7-fold decreases in inhibition potency, respectively, compared with that of CYP2B6 by sibutramine. sibutramine 128-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 23777987-7 2013 In addition, the CYP2B6 inhibitory potential of sibutramine was enhanced at a lower microsomal protein concentration of 0.05mg/ml. sibutramine 48-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 23777987-9 2013 These observations suggest that sibutramine is a selective and potent inhibitor of CYP2B6 in vitro, whereas inhibition of other CYPs is substantially lower. sibutramine 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 23777987-10 2013 These in vitro data support the use of sibutramine as a well-known inhibitor of CYP2B6 for routine screening of P450 reversible inhibition when human liver microsomes are used as the enzyme source. sibutramine 39-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 24005963-4 2013 Honokiol also moderately inhibited CYP2B6-mediated bupropion hydroxylation and CYP2D6-mediated bufuralol 1"-hydroxylation with K(i) values of 17.5 and 12.0 muM, respectively. Bupropion 51-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 23821647-1 2013 Cytochrome P450 (P450)-catalyzed oxidation of the aromatic ring of estradiol can result in 2- or 4-hydroxylation. Estradiol 67-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 24517233-3 2013 The CYP2B6 516TT genotype and male gender were significantly associated with occurrence of Efavirenz induced central nervous system disorders (OR 20.58, p=0.004) and the ABCB1 rs10276036TT genotype with Nevirapine induced skin hypersensitivity (OR 4.01, p=0.04). Nevirapine 203-213 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 23687222-1 2013 OBJECTIVE: Nevirapine is metabolized by cytochrome P450 (CYP) 2B6 and CYP3A4. Nevirapine 11-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-65 23639433-0 2013 Ethanol self-administration and nicotine treatment induce brain levels of CYP2B6 and CYP2E1 in African green monkeys. Ethanol 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 23639433-0 2013 Ethanol self-administration and nicotine treatment induce brain levels of CYP2B6 and CYP2E1 in African green monkeys. Nicotine 32-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 23639433-3 2013 The objective of this study was to determine the effects of ethanol self-administration and nicotine treatment, alone and in combination, on brain CYP2B6 and CYP2E1 levels in monkeys. Ethanol 60-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 23639433-3 2013 The objective of this study was to determine the effects of ethanol self-administration and nicotine treatment, alone and in combination, on brain CYP2B6 and CYP2E1 levels in monkeys. Nicotine 92-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 23639433-6 2013 Immunocytochemistry revealed induction of both CYP2B6 and CYP2E1 protein in certain brain regions and cells within monkey brain as a result of ethanol self-administration, nicotine treatment and combined exposure to both drugs. Ethanol 143-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 23639433-6 2013 Immunocytochemistry revealed induction of both CYP2B6 and CYP2E1 protein in certain brain regions and cells within monkey brain as a result of ethanol self-administration, nicotine treatment and combined exposure to both drugs. Nicotine 172-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 23639433-7 2013 Immunoblotting analyses demonstrated CYP2B6 induction by ethanol in the caudate, putamen and cerebellum (1.5-3.2 fold, P < 0.05), and CYP2E1 induction by nicotine in the frontal cortex and putamen (1.6-2.0 fold, P < 0.05). Ethanol 57-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 23639433-8 2013 Combined ethanol and nicotine exposure induced CYP2B6 in the caudate, putamen, thalamus and cerebellum (1.4-2.4 fold, P < 0.05), and CYP2E1 in the frontal cortex and putamen (1.5-1.8, P < 0.05). Ethanol 9-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 23639433-8 2013 Combined ethanol and nicotine exposure induced CYP2B6 in the caudate, putamen, thalamus and cerebellum (1.4-2.4 fold, P < 0.05), and CYP2E1 in the frontal cortex and putamen (1.5-1.8, P < 0.05). Nicotine 21-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 23639433-10 2013 In summary, ethanol and nicotine can induce CYP2B6 and CYP2E1 protein in the primate brain, which could potentially result in altered sensitivity to centrally acting drugs and toxins. Ethanol 12-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 23639433-10 2013 In summary, ethanol and nicotine can induce CYP2B6 and CYP2E1 protein in the primate brain, which could potentially result in altered sensitivity to centrally acting drugs and toxins. Nicotine 24-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 23905516-0 2013 Investigations of heme ligation and ligand switching in cytochromes p450 and p420. Heme 18-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 23905516-1 2013 It is generally accepted that the inactive P420 form of cytochrome P450 (CYP) involves the protonation of the native cysteine thiolate to form a neutral thiol heme ligand. cysteine thiolate 117-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-71 23905516-1 2013 It is generally accepted that the inactive P420 form of cytochrome P450 (CYP) involves the protonation of the native cysteine thiolate to form a neutral thiol heme ligand. thiol heme 153-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-71 23905516-2 2013 On the other hand, it has also been suggested that recruitment of a histidine to replace the native cysteine thiolate ligand might underlie the P450 P420 transition. Histidine 68-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-148 23905516-2 2013 On the other hand, it has also been suggested that recruitment of a histidine to replace the native cysteine thiolate ligand might underlie the P450 P420 transition. cysteine thiolate 100-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-148 23850985-3 2013 We found that both sesquiterpenes (1-30 muM) greatly induced expression of CYP2B6 and CYP3A4 but not CYP1A2 mRNAs in human primary hepatocytes (HPHs). Sesquiterpenes 19-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 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 2 subfamily B member 6 Homo sapiens 125-131 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 2 subfamily B member 6 Homo sapiens 125-131 23687222-10 2013 CONCLUSIONS: The effects on plasma drug exposure following single-dose nevirapine may be greater for CYP2B6 983T C than for 516G T and are less pronounced than at steady state. Nevirapine 71-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 23441978-3 2013 Artemether and lumefantrine are primarily metabolized by CYP3A; artemether is also metabolized to a lesser extent by CYP2B6, CYP2C9 and CYP2C19. Artemether 64-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 23709224-0 2013 Isolation and characterization of two geometric allene oxide isomers synthesized from 9S-hydroperoxylinoleic acid by cytochrome P450 CYP74C3: stereochemical assignment of natural fatty acid allene oxides. allene oxide 48-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 22634058-8 2013 The correlations between IC50 values obtained from the N-in-one and single substrate approaches were highly significant for CYP2Cs (r(2)=0.94), CYP3A4, omeprazole-sulfoxidation, (r(2)=0.89), followed by CYP1A2 and CYP2B6 (r(2)=0.82), and CYP2D6 (r(2)=0.80). n-in-one 55-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 214-220 22634058-4 2013 Chlorpyrifos and fenitrothion were most effective in inhibiting CYP1A1/2, and CYP2B6. Chlorpyrifos 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 22634058-4 2013 Chlorpyrifos and fenitrothion were most effective in inhibiting CYP1A1/2, and CYP2B6. Fenitrothion 17-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 24180002-0 2013 Effects of rifampin-based antituberculosis therapy on plasma efavirenz concentrations in children vary by CYP2B6 genotype. Rifampin 11-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 24180002-2 2013 However, rifampin, a key component of antituberculosis therapy, induces CYP2B6. Rifampin 9-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 23709224-0 2013 Isolation and characterization of two geometric allene oxide isomers synthesized from 9S-hydroperoxylinoleic acid by cytochrome P450 CYP74C3: stereochemical assignment of natural fatty acid allene oxides. 9s-hydroperoxylinoleic acid 86-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 23709224-0 2013 Isolation and characterization of two geometric allene oxide isomers synthesized from 9S-hydroperoxylinoleic acid by cytochrome P450 CYP74C3: stereochemical assignment of natural fatty acid allene oxides. natural fatty acid 171-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 23709224-0 2013 Isolation and characterization of two geometric allene oxide isomers synthesized from 9S-hydroperoxylinoleic acid by cytochrome P450 CYP74C3: stereochemical assignment of natural fatty acid allene oxides. allene oxides 190-203 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 23709224-1 2013 Specialized cytochromes P450 or catalase-related hemoproteins transform fatty acid hydroperoxides to allene oxides, highly reactive epoxides leading to cyclopentenones and other products. Lipid Peroxides 72-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 23709224-1 2013 Specialized cytochromes P450 or catalase-related hemoproteins transform fatty acid hydroperoxides to allene oxides, highly reactive epoxides leading to cyclopentenones and other products. allene oxides 101-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 23709224-1 2013 Specialized cytochromes P450 or catalase-related hemoproteins transform fatty acid hydroperoxides to allene oxides, highly reactive epoxides leading to cyclopentenones and other products. Epoxy Compounds 132-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 23709224-1 2013 Specialized cytochromes P450 or catalase-related hemoproteins transform fatty acid hydroperoxides to allene oxides, highly reactive epoxides leading to cyclopentenones and other products. cyclopentenone 152-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 23733841-9 2013 RESULTS: Methadone (10 microM) increased methadone N-demethylation 2-fold, CYP2B6 and CYP3A4 mRNA 3-fold, and protein expression 2-fold. Methadone 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 23733841-3 2013 Although methadone N-demethylation is catalyzed in vitro by cytochrome P4502B6 (CYP2B6) and CYP3A4, and clearance in vivo depends on CYP2B6, mechanism(s) of autoinduction are incompletely understood. Methadone 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 23786449-0 2013 Structural and thermodynamic basis of (+)-alpha-pinene binding to human cytochrome P450 2B6. alpha-pinene 38-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-91 23880855-4 2013 Ospemifene and its main metabolites 4-hydroxyospemifene and 4"-hydroxyospemifene weakly inhibited a number of CYPs (CYP2B6, CYP2C9, CYP2C19, CYP2C8, and CYP2D6) in vitro. Ospemifene 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 23880855-4 2013 Ospemifene and its main metabolites 4-hydroxyospemifene and 4"-hydroxyospemifene weakly inhibited a number of CYPs (CYP2B6, CYP2C9, CYP2C19, CYP2C8, and CYP2D6) in vitro. 4-hydroxyospemifene 36-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 23880855-4 2013 Ospemifene and its main metabolites 4-hydroxyospemifene and 4"-hydroxyospemifene weakly inhibited a number of CYPs (CYP2B6, CYP2C9, CYP2C19, CYP2C8, and CYP2D6) in vitro. 4-hydroxyospemifene 60-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 23733841-3 2013 Although methadone N-demethylation is catalyzed in vitro by cytochrome P4502B6 (CYP2B6) and CYP3A4, and clearance in vivo depends on CYP2B6, mechanism(s) of autoinduction are incompletely understood. Methadone 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 23733841-16 2013 CONCLUSIONS: Methadone caused concentration-dependent autoinduction of methadone N-demethylation in human hepatocytes, related to induction of CYP2B6 and CYP3A4 mRNA expression, protein expression, and catalytic activity. Methadone 13-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 23774830-6 2013 The enzyme kinetic studies showed that the initial metabolic step in humans, the N-deethylation, was catalyzed by CYP2B6 and CYP3A4. Nitrogen 81-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 23548939-9 2013 This was associated with a reduction in the P450 aromatase and P450scc (cholesterol side-chain cleavage cytochromes P450) (P450scc) protein levels but not those of 3beta-hydroxysteroid dehydrogenase (3beta-HSD) or steroidogenic acute regulatory protein (StAR) and with a decrease in IGF-I-induced IGF-I receptor and mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation. Cholesterol 72-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-121 21790905-0 2013 CYP2B6 SNPs are associated with methadone dose required for effective treatment of opioid addiction. Methadone 32-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 21790905-3 2013 Methadone metabolism is attributed primarily to cytochrome P450 enzymes CYP3A4, CYP2B6 and CYP2D6. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 21790905-4 2013 The CYP2B6*6 allele [single nucleotide polymorphisms (SNPs) 785A>G (rs2279343) and 516G>T (rs3745274)] was associated with slow methadone metabolism. Methadone 134-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 21790905-5 2013 To explore the effects of CYP2B6*6 allele on methadone dose requirement, it was genotyped in a well-characterized sample of 74 Israeli former heroin addicts in MMT. Methadone 45-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 21790905-5 2013 To explore the effects of CYP2B6*6 allele on methadone dose requirement, it was genotyped in a well-characterized sample of 74 Israeli former heroin addicts in MMT. mmt 160-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 21790905-9 2013 The mean methadone doses required by subjects homozygous for the variant alleles of the CYP2B6 SNPs 785A>G and 516G>T (88, 96mg, respectively) were significantly lower than those of the heterozygotes (133, 129mg, respectively) and the non-carriers (150, 151mg, respectively) (nominal P=0.012, 0.048, respectively). Methadone 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 23859571-5 2013 A preliminary analysis confirmed that CYP2B6*6 (516G>T and 785A>G) was the most highly correlated (p = 0.005) with AEs and high plasma concentrations. aes 121-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 24043992-0 2013 Frequencies of Cytochrome P450 2B6 and 2C8 Allelic Variants in the Mozambican Population. mozambican 67-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-42 24043992-5 2013 This study aimed to analyse the frequencies of allelic variants of CYP2B6 and CYP2C8 in the Mozambican population. mozambican 92-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 24043992-10 2013 CONCLUSION: The frequencies of the allelic variants of the CYP2B6 and CYP2C8 genes were found to be homogeneously distributed in the Mozambican population and were comparable to other African populations. mozambican 133-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 23592585-0 2013 How is a metabolic intermediate formed in the mechanism-based inactivation of cytochrome P450 by using 1,1-dimethylhydrazine: hydrogen abstraction or nitrogen oxidation? dimazine 103-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-93 23615745-0 2013 Cytochrome P450 2B6 and 2C9 genotype polymorphism--a possible cause of prasugrel low responsiveness. Prasugrel Hydrochloride 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-27 23615745-16 2013 In conclusion, CYP2B6 and CYP2C9 polymorphisms seem to be associated with prasugrel low-response. Prasugrel Hydrochloride 74-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 23840296-0 2013 Genetic Variants of Pregnane X Receptor (PXR) and CYP2B6 Affect the Induction of Bupropion Hydroxylation by Sodium Ferulate. Bupropion 81-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 23840296-0 2013 Genetic Variants of Pregnane X Receptor (PXR) and CYP2B6 Affect the Induction of Bupropion Hydroxylation by Sodium Ferulate. ferulic acid 108-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 23840296-1 2013 UNLABELLED: This study investigated the effects of pregnane X receptor (PXR/NR1I2) and CYP2B6 genetic variants on sodium ferulate (SF)-mediated induction of bupropion hydroxylation. ferulic acid 114-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 23840296-1 2013 UNLABELLED: This study investigated the effects of pregnane X receptor (PXR/NR1I2) and CYP2B6 genetic variants on sodium ferulate (SF)-mediated induction of bupropion hydroxylation. ferulic acid 131-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 23840296-8 2013 In conclusion, it is suggested that NR1I2 variants decrease the bupropion hydroxylation induced by SF treatment, particularly in CYP2B6*6 carriers. Bupropion 64-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 23592585-0 2013 How is a metabolic intermediate formed in the mechanism-based inactivation of cytochrome P450 by using 1,1-dimethylhydrazine: hydrogen abstraction or nitrogen oxidation? Hydrogen 126-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-93 23592585-0 2013 How is a metabolic intermediate formed in the mechanism-based inactivation of cytochrome P450 by using 1,1-dimethylhydrazine: hydrogen abstraction or nitrogen oxidation? Nitrogen 150-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-93 23592585-4 2013 We used density functional theory (DFT) calculations to compare four possible mechanisms underlying the reaction between 1,1-dimethylhydrazine (or unsymmetrical dimethylhydrazine, UDMH) and the reactive compound I (Cpd I) intermediate of P450. dimazine 121-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 238-242 23592585-4 2013 We used density functional theory (DFT) calculations to compare four possible mechanisms underlying the reaction between 1,1-dimethylhydrazine (or unsymmetrical dimethylhydrazine, UDMH) and the reactive compound I (Cpd I) intermediate of P450. Dimethylhydrazines 125-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 238-242 23550066-0 2013 The CYP2B6*6 allele significantly alters the N-demethylation of ketamine enantiomers in vitro. Nitrogen 45-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 23550066-0 2013 The CYP2B6*6 allele significantly alters the N-demethylation of ketamine enantiomers in vitro. Ketamine 64-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 23539296-4 2013 In the present study, we found that activation of p38 MAPK by anisomycin potentiated induction of CYP2B6 mRNA by CAR ligand in HepG2 cells to levels observed in ligand-treated human primary hepatocytes. Anisomycin 62-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 23550066-1 2013 Ketamine is primarily metabolized to norketamine by hepatic CYP2B6 and CYP3A4-mediated N-demethylation. Ketamine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 23539296-5 2013 siRNA knockdown of p38 MAPK abrogated the ability of anisomycin to synergistically induce CYP2B6 mRNA. Anisomycin 53-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 23242004-1 2013 PURPOSE: Tramadol is mainly metabolized by the cytochrome P450 (CYP) 2D6, CYP2B6 and CYP3A4 enzymes. Tramadol 9-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 23550066-1 2013 Ketamine is primarily metabolized to norketamine by hepatic CYP2B6 and CYP3A4-mediated N-demethylation. norketamine 37-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 23550066-3 2013 The CYP2B6*6 allele is associated with reduced enzyme expression and activity that may lead to interindividual variability in ketamine metabolism. Ketamine 126-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 23550066-8 2013 The intrinsic clearance for both ketamine enantiomers by the high-affinity enzyme in HLMs with CYP2B6*1/*1 genotype were at least 2-fold and 6-fold higher, respectively, than those for CYP2B6*1/*6 genotype and CYP2B6*6/*6 genotype. Ketamine 33-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-101 23550066-8 2013 The intrinsic clearance for both ketamine enantiomers by the high-affinity enzyme in HLMs with CYP2B6*1/*1 genotype were at least 2-fold and 6-fold higher, respectively, than those for CYP2B6*1/*6 genotype and CYP2B6*6/*6 genotype. Ketamine 33-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 185-191 23550066-8 2013 The intrinsic clearance for both ketamine enantiomers by the high-affinity enzyme in HLMs with CYP2B6*1/*1 genotype were at least 2-fold and 6-fold higher, respectively, than those for CYP2B6*1/*6 genotype and CYP2B6*6/*6 genotype. Ketamine 33-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 185-191 23550066-12 2013 These results indicate a major role of CYP2B6 in ketamine N-demethylation in vitro and a significant impact of the CYP2B6*6 allele on enzyme-ketamine binding and catalytic activity. Ketamine 49-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 23550066-12 2013 These results indicate a major role of CYP2B6 in ketamine N-demethylation in vitro and a significant impact of the CYP2B6*6 allele on enzyme-ketamine binding and catalytic activity. Ketamine 141-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 23298862-0 2013 Methadone N-demethylation by the common CYP2B6 allelic variant CYP2B6.6. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 23369276-3 2013 Site-directed mutagenesis was conducted to produce a mutant P450 that could attach biotin site-specifically. Biotin 83-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-64 23369276-5 2013 Ultraviolet-visible spectroscopy of the carbon monoxide-bound P450, circular dichroism data, and the ratio of the active form to the sum of the active form and the inactive form indicated that this decrease in activity was because of a conformational change in the tertiary structure surrounding the active center after avidin binding, while the secondary structure of P450 remained unchanged. Carbon Monoxide 40-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 23524664-0 2013 Influence of the CYP2B6 polymorphism on the pharmacokinetics of mitotane. Mitotane 64-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 23524664-1 2013 OBJECTIVE: The aim of this study was to assess the potential impact of the pharmacogenetic variability of CYP2B6 and ABCB1 genes on the pharmacokinetics of mitotane. Mitotane 156-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 23524664-5 2013 Multivariate logistic regression analysis showed that only the CYP2B6 rs3745274GT/TT genotype (odds ratio=10.7; P=0.017) was a predictor of mitotane plasma concentrations of at least 14 microg/ml after 3 months of treatment. Mitotane 140-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 23524664-7 2013 CONCLUSION: Evaluation of the CYP2B6 polymorphism enabled prediction of the individual response to adjuvant mitotane treatment. Mitotane 108-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 23537005-0 2013 Biotransformation of 2,2",4,4"-tetrabromodiphenyl ether (BDE-47) by human liver microsomes: identification of cytochrome P450 2B6 as the major enzyme involved. 2,2',4,4'-tetrabromodiphenyl ether 21-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-129 23537005-0 2013 Biotransformation of 2,2",4,4"-tetrabromodiphenyl ether (BDE-47) by human liver microsomes: identification of cytochrome P450 2B6 as the major enzyme involved. 2,2',4,4'-tetrabromodiphenyl ether 57-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-129 23330950-1 2013 INTRODUCTION: Cytochromes P450 (P450) and associated monooxygenases are a family of heme proteins involved in metabolism of endogenous compounds (arachidonic acid, eicosanoids and prostaglandins) as also xenobiotics including drugs and environmental chemicals. Arachidonic Acid 146-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 23330950-1 2013 INTRODUCTION: Cytochromes P450 (P450) and associated monooxygenases are a family of heme proteins involved in metabolism of endogenous compounds (arachidonic acid, eicosanoids and prostaglandins) as also xenobiotics including drugs and environmental chemicals. Arachidonic Acid 146-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 23330950-1 2013 INTRODUCTION: Cytochromes P450 (P450) and associated monooxygenases are a family of heme proteins involved in metabolism of endogenous compounds (arachidonic acid, eicosanoids and prostaglandins) as also xenobiotics including drugs and environmental chemicals. Eicosanoids 164-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 23330950-1 2013 INTRODUCTION: Cytochromes P450 (P450) and associated monooxygenases are a family of heme proteins involved in metabolism of endogenous compounds (arachidonic acid, eicosanoids and prostaglandins) as also xenobiotics including drugs and environmental chemicals. Eicosanoids 164-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 23330950-1 2013 INTRODUCTION: Cytochromes P450 (P450) and associated monooxygenases are a family of heme proteins involved in metabolism of endogenous compounds (arachidonic acid, eicosanoids and prostaglandins) as also xenobiotics including drugs and environmental chemicals. Prostaglandins 180-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 23330950-1 2013 INTRODUCTION: Cytochromes P450 (P450) and associated monooxygenases are a family of heme proteins involved in metabolism of endogenous compounds (arachidonic acid, eicosanoids and prostaglandins) as also xenobiotics including drugs and environmental chemicals. Prostaglandins 180-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 23330950-5 2013 AREAS COVERED: An overview of P450-mediated metabolism in brain is presented focusing on distinct differences seen in expression of P450 enzymes, generation of unique P450 enzymes in brain through alternate splicing and their consequences in terms of metabolism of psychoactive drugs and inflammatory prompts, such as leukotrienes, thus modulating inflammatory response. Leukotrienes 318-330 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 23381968-1 2013 In this study, we assessed the effects of clopidogrel and clarithromycin, known CYP2B6 and CYP3A inhibitors, respectively, on the enantioselective disposition of racemic sibutramine in conjunction with CYP2B6 polymorphisms in humans. sibutramine 170-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 23381968-5 2013 Exposure to sibutramine was higher in subjects with the CYP2B6*6/*6 genotype, but no statistical difference was observed among the CYP2B6 genotypes. sibutramine 12-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 23381968-6 2013 These results suggest that the enantioselective disposition of sibutramine and its active metabolites are influenced by the altered genetic and environmental factors of CYP2B6 and CYP3A activity in vivo. sibutramine 63-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 23298862-13 2013 Diminished methadone N-demethylation by CYP2B6.6 may provide a mechanistic explanation for clinical observations of altered methadone disposition in individuals carrying the CYP2B6*6 polymorphism. Nitrogen 21-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 174-180 23298862-13 2013 Diminished methadone N-demethylation by CYP2B6.6 may provide a mechanistic explanation for clinical observations of altered methadone disposition in individuals carrying the CYP2B6*6 polymorphism. Methadone 124-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 23298862-13 2013 Diminished methadone N-demethylation by CYP2B6.6 may provide a mechanistic explanation for clinical observations of altered methadone disposition in individuals carrying the CYP2B6*6 polymorphism. Methadone 124-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 174-180 23604525-6 2013 Clomiphene, tamoxifen and toremifene were biotransformed to 22, 23 and 18 metabolites respectively, these phase I reactions being catalyzed mainly by CYP3A4 and CYP2D6 isoforms and, to a lesser degree, by CYP3A5, CYP2B6, CYP2C9, CYP2C19 isoforms. Clomiphene 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 213-219 23604525-6 2013 Clomiphene, tamoxifen and toremifene were biotransformed to 22, 23 and 18 metabolites respectively, these phase I reactions being catalyzed mainly by CYP3A4 and CYP2D6 isoforms and, to a lesser degree, by CYP3A5, CYP2B6, CYP2C9, CYP2C19 isoforms. Tamoxifen 12-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 213-219 23604525-6 2013 Clomiphene, tamoxifen and toremifene were biotransformed to 22, 23 and 18 metabolites respectively, these phase I reactions being catalyzed mainly by CYP3A4 and CYP2D6 isoforms and, to a lesser degree, by CYP3A5, CYP2B6, CYP2C9, CYP2C19 isoforms. Toremifene 26-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 213-219 23591849-0 2013 Pilot study of CYP2B6 genetic variation to explore the contribution of nitrosamine activation to lung carcinogenesis. Nitrosamines 71-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 23591849-1 2013 We explored the contribution of nitrosamine metabolism to lung cancer in a pilot investigation of genetic variation in CYP2B6, a high-affinity enzymatic activator of tobacco-specific nitrosamines with a negligible role in nicotine metabolism. Nitrosamines 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 23591849-1 2013 We explored the contribution of nitrosamine metabolism to lung cancer in a pilot investigation of genetic variation in CYP2B6, a high-affinity enzymatic activator of tobacco-specific nitrosamines with a negligible role in nicotine metabolism. Nitrosamines 183-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 23591849-1 2013 We explored the contribution of nitrosamine metabolism to lung cancer in a pilot investigation of genetic variation in CYP2B6, a high-affinity enzymatic activator of tobacco-specific nitrosamines with a negligible role in nicotine metabolism. Nicotine 222-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 23591849-7 2013 Findings from this pilot point to genetic variation in CYP2B6 as a lung cancer risk factor supporting a role for nitrosamine metabolic activation in the molecular mechanism of lung carcinogenesis. Nitrosamines 113-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 23415833-7 2013 The Vmax for BCP formation was 47.9, 25.1, and 19.2 nmol/min/nmol CYP for CYP2B6, 2C19, and 3A4, respectively. 4-bromo-2-chlorophenol 13-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 23415833-8 2013 Intrinsic clearance (Vmax/Km) values of 48.8, 46.9, and 1.02 ml/min/nmol CYP 2C19, 2B6, and 3A4, respectively, indicate that CYP2C19 and CYP2B6 are primarily responsible for the detoxification of profenofos. profenofos 196-206 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 23298862-0 2013 Methadone N-demethylation by the common CYP2B6 allelic variant CYP2B6.6. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 23298862-2 2013 Methadone metabolism clinically occurs primarily by N-demethylation to 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP), catalyzed predominantly by CYP2B6. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 23298862-2 2013 Methadone metabolism clinically occurs primarily by N-demethylation to 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP), catalyzed predominantly by CYP2B6. 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine 71-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 23298862-2 2013 Methadone metabolism clinically occurs primarily by N-demethylation to 2-ethyl-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP), catalyzed predominantly by CYP2B6. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 117-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 23298862-3 2013 Retrospective studies suggest that the common allele variant CYP2B6*6 may influence methadone plasma concentrations. Methadone 84-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 23298862-5 2013 This investigation compared methadone N-demethylation by CYP2B6.6 with that by wild-type CYP2B6.1. Methadone 28-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 23298862-6 2013 Methadone enantiomer and racemate N-demethylation by recombinant-expressed CYP2B6.6 and CYP2B6.1 was determined. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 23298862-6 2013 Methadone enantiomer and racemate N-demethylation by recombinant-expressed CYP2B6.6 and CYP2B6.1 was determined. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 23298862-6 2013 Methadone enantiomer and racemate N-demethylation by recombinant-expressed CYP2B6.6 and CYP2B6.1 was determined. Nitrogen 34-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 23298862-6 2013 Methadone enantiomer and racemate N-demethylation by recombinant-expressed CYP2B6.6 and CYP2B6.1 was determined. Nitrogen 34-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 23298862-7 2013 At substrate concentrations (0.25-2 microM) approximating plasma concentrations occurring clinically, rates of methadone enantiomer N-demethylation by CYP2B6.6, incubated individually or as the racemate, were one-third to one-fourth those by CYP2B6.1. Methadone 111-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 23298862-7 2013 At substrate concentrations (0.25-2 microM) approximating plasma concentrations occurring clinically, rates of methadone enantiomer N-demethylation by CYP2B6.6, incubated individually or as the racemate, were one-third to one-fourth those by CYP2B6.1. Methadone 111-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-248 23298862-8 2013 For methadone individual enantiomers and metabolism by CYP2B6.6 compared with CYP2B6.1, Vmax was diminished, Ks was greater and the in vitro intrinsic clearance was diminished 5- to 6-fold. Methadone 4-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 23298862-8 2013 For methadone individual enantiomers and metabolism by CYP2B6.6 compared with CYP2B6.1, Vmax was diminished, Ks was greater and the in vitro intrinsic clearance was diminished 5- to 6-fold. Methadone 4-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 23298862-8 2013 For methadone individual enantiomers and metabolism by CYP2B6.6 compared with CYP2B6.1, Vmax was diminished, Ks was greater and the in vitro intrinsic clearance was diminished 5- to 6-fold. Potassium 109-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 23298862-8 2013 For methadone individual enantiomers and metabolism by CYP2B6.6 compared with CYP2B6.1, Vmax was diminished, Ks was greater and the in vitro intrinsic clearance was diminished 5- to 6-fold. Potassium 109-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 23298862-10 2013 Both CYP2B6.6 and CYP2B6.1 showed similar stereoselectivity (S>R-methadone). Methadone 66-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-11 23298862-10 2013 Both CYP2B6.6 and CYP2B6.1 showed similar stereoselectivity (S>R-methadone). Methadone 66-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 23298862-11 2013 Human liver microsomes with diminished CYP2B6 content due to a CYP2B6*6 allele had lower rates of methadone N-demethylation. Methadone 98-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 23298862-11 2013 Human liver microsomes with diminished CYP2B6 content due to a CYP2B6*6 allele had lower rates of methadone N-demethylation. Methadone 98-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 23298862-12 2013 Results show that methadone N-demethylation catalyzed by CYP2B6.6, the CYP2B6 variant encoded by the CYP2B6*6 polymorphism, is catalytically deficient compared with wild-type CYP2B6.1. Methadone 18-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 23298862-12 2013 Results show that methadone N-demethylation catalyzed by CYP2B6.6, the CYP2B6 variant encoded by the CYP2B6*6 polymorphism, is catalytically deficient compared with wild-type CYP2B6.1. Methadone 18-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 23298862-12 2013 Results show that methadone N-demethylation catalyzed by CYP2B6.6, the CYP2B6 variant encoded by the CYP2B6*6 polymorphism, is catalytically deficient compared with wild-type CYP2B6.1. Methadone 18-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 23298862-12 2013 Results show that methadone N-demethylation catalyzed by CYP2B6.6, the CYP2B6 variant encoded by the CYP2B6*6 polymorphism, is catalytically deficient compared with wild-type CYP2B6.1. Methadone 18-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 23298862-13 2013 Diminished methadone N-demethylation by CYP2B6.6 may provide a mechanistic explanation for clinical observations of altered methadone disposition in individuals carrying the CYP2B6*6 polymorphism. Methadone 11-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 23298862-13 2013 Diminished methadone N-demethylation by CYP2B6.6 may provide a mechanistic explanation for clinical observations of altered methadone disposition in individuals carrying the CYP2B6*6 polymorphism. Methadone 11-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 174-180 23298862-13 2013 Diminished methadone N-demethylation by CYP2B6.6 may provide a mechanistic explanation for clinical observations of altered methadone disposition in individuals carrying the CYP2B6*6 polymorphism. Nitrogen 21-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 23249762-8 2013 In kinetic studies of BDE-47 metabolism by CYP2B6 and pooled HLMs, we found Km values ranging from 3.8 to 6.4 microM and 7.0 to 11.4 microM, respectively, indicating the high affinity toward the formation of OH-BDEs. oh-bdes 208-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 23249762-6 2013 RESULTS: CYP2B6 was the predominant CYP capable of forming six OH-BDEs, including 3-OH-BDE-47, 5-OH-BDE-47, 6-OH-BDE-47, 4-OH-BDE-42, 4 -OH-BDE-49, and a metabolite tentatively identified as 2 -OH-BDE-66. oh-bdes 63-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 23249762-9 2013 CONCLUSION: Our findings support a predominant role of CYP2B6 in the metabolism of BDE-47 to potentially toxic metabolites, including a hypothesized di-OH-tetrabrominated dioxin metabolite. di-oh 149-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 23099620-1 2013 PURPOSE: We assessed possible drug interactions of tramadol given concomitantly with the potent CYP2B6 inhibitor ticlopidine, alone or together with the potent CYP3A4 and P-glycoprotein inhibitor itraconazole. Ticlopidine 113-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 23249762-9 2013 CONCLUSION: Our findings support a predominant role of CYP2B6 in the metabolism of BDE-47 to potentially toxic metabolites, including a hypothesized di-OH-tetrabrominated dioxin metabolite. Dioxins 171-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 23099620-7 2013 CONCLUSIONS: Ticlopidine increased exposure to tramadol, reduced its renal clearance and inhibited the formation of M1, most likely via inhibition of CYP2B6 and/or CYP2D6. Ticlopidine 13-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 23467454-3 2013 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. artemisinin 43-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 27536444-5 2013 METHODS: The ability of 4-AP to inhibit CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4/5 in a direct and time-dependent manner was evaluated using pooled human liver microsomes. 4-Aminopyridine 24-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 23858795-9 2013 These results suggest that 3"-OH-CB146 is formed by PB-inducible cytochrome P450 (CYP2B enzymes) in animal and human livers and 4-OH-CB146 is a major metabolite in rat and human liver. 3"-oh-cb146 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-87 23858795-9 2013 These results suggest that 3"-OH-CB146 is formed by PB-inducible cytochrome P450 (CYP2B enzymes) in animal and human livers and 4-OH-CB146 is a major metabolite in rat and human liver. Phenobarbital 52-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-87 23153057-3 2013 The cocktail strategy (in situ incubation) was used to assess the induction of CYP1A2, CYP2B6, CYP2C9 and CYP3A4 by using the recommended probe substrate, i.e. phenacetin, bupropion, diclofenac and midazolam, respectively. Bupropion 172-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 23153057-3 2013 The cocktail strategy (in situ incubation) was used to assess the induction of CYP1A2, CYP2B6, CYP2C9 and CYP3A4 by using the recommended probe substrate, i.e. phenacetin, bupropion, diclofenac and midazolam, respectively. Diclofenac 183-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 23153057-3 2013 The cocktail strategy (in situ incubation) was used to assess the induction of CYP1A2, CYP2B6, CYP2C9 and CYP3A4 by using the recommended probe substrate, i.e. phenacetin, bupropion, diclofenac and midazolam, respectively. Midazolam 198-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 23467454-3 2013 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. Bupropion 56-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 23467454-3 2013 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. Cyclophosphamide 67-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 23467454-3 2013 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. efavirenz 85-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 23467454-3 2013 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. Ketamine 96-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 23467454-3 2013 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. Methadone 110-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 23238783-0 2013 The influence of sex, ethnicity, and CYP2B6 genotype on bupropion metabolism as an index of hepatic CYP2B6 activity in humans. Bupropion 56-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 23238783-0 2013 The influence of sex, ethnicity, and CYP2B6 genotype on bupropion metabolism as an index of hepatic CYP2B6 activity in humans. Bupropion 56-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 23238783-10 2013 These results suggest that CYP2B6 genotype is the most important patient variable for predicting the level of CYP2B6 activity in women, when measured by the metabolism of bupropion. Bupropion 171-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 23238783-10 2013 These results suggest that CYP2B6 genotype is the most important patient variable for predicting the level of CYP2B6 activity in women, when measured by the metabolism of bupropion. Bupropion 171-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 23238783-11 2013 The bupropion metabolic ratio appears to detect known differences in CYP2B6 activity associated with genetic polymorphism, across different ethnic groups. Bupropion 4-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 23344581-1 2013 BACKGROUND: Bupropion, an antidepressant and smoking cessation medication, is metabolized to hydroxybupropion (HB), an active metabolite, primarily by CYP2B6. Bupropion 12-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 23826879-7 2013 Amitifadine was a moderate inhibitor of the human isoforms of the major drug metabolizing enzymes CYP2D6, CYP3A4, CYP2C9, and CYP2C19 (IC50 = 9 - 100 muM), but was a potent inhibitor of human CYP2B6 (IC50 = 1.8 muM). 1-(3,4-dichlorophenyl)-3-azabicyclo-(3.1.0)hexane hydrochloride 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 23361846-0 2013 Role of cytochrome P4502B6 in methadone metabolism and clearance. Methadone 30-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-26 23361846-1 2013 Methadone N-demethylation in vitro is catalyzed by hepatic cytochrome P4502B6 (CYP2B6) and CYP3A4, but clinical disposition is often attributed to CYP3A4. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-77 23361846-1 2013 Methadone N-demethylation in vitro is catalyzed by hepatic cytochrome P4502B6 (CYP2B6) and CYP3A4, but clinical disposition is often attributed to CYP3A4. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 23361846-2 2013 This investigation tested the hypothesis that CYP2B6 is a prominent CYP isoform responsible for clinical methadone N-demethylation and clearance, using the in vivo mechanism-based CYP2B6 inhibitor ticlopidine, given orally for 4 days. Methadone 105-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 23361846-2 2013 This investigation tested the hypothesis that CYP2B6 is a prominent CYP isoform responsible for clinical methadone N-demethylation and clearance, using the in vivo mechanism-based CYP2B6 inhibitor ticlopidine, given orally for 4 days. Ticlopidine 197-208 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 23361846-7 2013 CYP2B6 inhibition reduces methadone N-demethylation and clearance, and alters methadone concentrations, demonstrating an important role for CYP2B6 in clinical methadone disposition. Methadone 26-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23361846-7 2013 CYP2B6 inhibition reduces methadone N-demethylation and clearance, and alters methadone concentrations, demonstrating an important role for CYP2B6 in clinical methadone disposition. Nitrogen 36-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23361846-7 2013 CYP2B6 inhibition reduces methadone N-demethylation and clearance, and alters methadone concentrations, demonstrating an important role for CYP2B6 in clinical methadone disposition. Methadone 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23361846-7 2013 CYP2B6 inhibition reduces methadone N-demethylation and clearance, and alters methadone concentrations, demonstrating an important role for CYP2B6 in clinical methadone disposition. Methadone 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 23361846-7 2013 CYP2B6 inhibition reduces methadone N-demethylation and clearance, and alters methadone concentrations, demonstrating an important role for CYP2B6 in clinical methadone disposition. Methadone 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23361846-7 2013 CYP2B6 inhibition reduces methadone N-demethylation and clearance, and alters methadone concentrations, demonstrating an important role for CYP2B6 in clinical methadone disposition. Methadone 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 25505649-2 2013 Artemether is a substrate of CYP3A4 and CYP2B6, antiretrovirals such as efavirenz induce these enzymes and have the potential to reduce artemether pharmacokinetic exposure. Artemether 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 25505649-2 2013 Artemether is a substrate of CYP3A4 and CYP2B6, antiretrovirals such as efavirenz induce these enzymes and have the potential to reduce artemether pharmacokinetic exposure. efavirenz 72-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 25505649-2 2013 Artemether is a substrate of CYP3A4 and CYP2B6, antiretrovirals such as efavirenz induce these enzymes and have the potential to reduce artemether pharmacokinetic exposure. Artemether 136-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 23344581-0 2013 Influence of CYP2B6 genetic variants on plasma and urine concentrations of bupropion and metabolites at steady state. Bupropion 75-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 23344581-1 2013 BACKGROUND: Bupropion, an antidepressant and smoking cessation medication, is metabolized to hydroxybupropion (HB), an active metabolite, primarily by CYP2B6. hydroxybupropion 93-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 23344581-2 2013 OBJECTIVES: To compare plasma concentrations of bupropion and metabolites at steady state in healthy volunteers with and without CYP2B6 genetic variants. Bupropion 48-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 23344581-6 2013 CONCLUSION: As HB is active and its steady-state concentrations are more than 10 times higher than bupropion, CYP2B6 variants are likely to affect pharmacological activity. Bupropion 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 23254426-0 2013 Impact of pharmacogenetic markers of CYP2B6, clinical factors, and drug-drug interaction on efavirenz concentrations in HIV/tuberculosis-coinfected patients. efavirenz 92-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 23245243-8 2013 Due to substantial inter-substrate interaction, chlorzoxazone (CYP2E1) and bupropion (CYP2B6) were removed from the initial seven probes CYP cocktail assay. Bupropion 75-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 23388178-3 2013 Following release from the phospholipid membrane, arachidonic acid can be metabolized into different classes of eicosanoids through cyclooxygenases, lipoxygenases, or p450 epoxygenase pathways. Phospholipids 27-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-171 23388178-3 2013 Following release from the phospholipid membrane, arachidonic acid can be metabolized into different classes of eicosanoids through cyclooxygenases, lipoxygenases, or p450 epoxygenase pathways. Arachidonic Acid 50-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-171 23388178-3 2013 Following release from the phospholipid membrane, arachidonic acid can be metabolized into different classes of eicosanoids through cyclooxygenases, lipoxygenases, or p450 epoxygenase pathways. Eicosanoids 112-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-171 22900583-2 2013 Ritonavir is a potent CYP3A inhibitor and was shown in vitro as a CYP2B6 inhibitor. Ritonavir 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 23249875-0 2013 Contribution of CYP2B6 alleles in explaining extreme (S)-methadone plasma levels: a CYP2B6 gene resequencing study. Methadone 53-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 23288240-0 2013 PXR polymorphisms interacted with CYP2B6 polymorphisms on methadone metabolites. Methadone 58-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 22815312-0 2013 Quantitative prediction of CYP2B6 induction by estradiol during pregnancy: potential explanation for increased methadone clearance during pregnancy. Estradiol 47-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 22815312-0 2013 Quantitative prediction of CYP2B6 induction by estradiol during pregnancy: potential explanation for increased methadone clearance during pregnancy. Methadone 111-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 22815312-4 2013 In rodent models and in HepG2 cells, CYP2B enzymes have been shown to be regulated by estradiol. Estradiol 86-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-42 22815312-5 2013 Because estradiol concentrations increase by ~50-fold during human pregnancy, it was hypothesized that the increasing estradiol concentrations during human pregnancy would result in induction of CYP2B6 activity. Estradiol 8-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-201 22815312-5 2013 Because estradiol concentrations increase by ~50-fold during human pregnancy, it was hypothesized that the increasing estradiol concentrations during human pregnancy would result in induction of CYP2B6 activity. Estradiol 118-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-201 22815312-8 2013 At 100 nM total estradiol, a concentration achievable during the third trimester of pregnancy, CYP2B6 activity was predicted to increase by 1.5-3-fold, based on increased CYP2B6 activity and mRNA. Estradiol 16-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-101 22815312-8 2013 At 100 nM total estradiol, a concentration achievable during the third trimester of pregnancy, CYP2B6 activity was predicted to increase by 1.5-3-fold, based on increased CYP2B6 activity and mRNA. Estradiol 16-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 22815312-9 2013 When the E(max) and EC(50) values were compared with those for carbamazepine and rifampin, estradiol was found to be as potent an inducer of CYP2B6 as rifampin and carbamazepine. Estradiol 91-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 22815312-10 2013 These data suggest that, during human pregnancy, the increasing estradiol concentrations will result in increased clearance of drugs that have CYP2B6-mediated clearance pathways. Estradiol 64-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 22837389-0 2013 Isoform-specific regulation of cytochromes P450 expression by estradiol and progesterone. Estradiol 62-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 22837389-0 2013 Isoform-specific regulation of cytochromes P450 expression by estradiol and progesterone. Progesterone 76-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 22837389-4 2013 The results showed that estradiol enhances CYP2A6, CYP2B6, and CYP3A4 expression, whereas progesterone induces CYP2A6, CYP2B6, CYP2C8, CYP3A4, and CYP3A5 expression. Estradiol 24-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 22837389-4 2013 The results showed that estradiol enhances CYP2A6, CYP2B6, and CYP3A4 expression, whereas progesterone induces CYP2A6, CYP2B6, CYP2C8, CYP3A4, and CYP3A5 expression. Progesterone 90-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 23249875-1 2013 BACKGROUND: (S)-Methadone, metabolized mainly by CYP2B6, shows a wide interindividual variability in its pharmacokinetics and pharmacodynamics. Methadone 12-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 23249875-2 2013 METHODS: Resequencing of the CYP2B6 gene was performed in 12 and 35 selected individuals with high (S)-methadone plasma exposure and low (S)-methadone plasma exposure, respectively, from a previously described cohort of 276 patients undergoing methadone maintenance treatment. Methadone 103-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 23249875-2 2013 METHODS: Resequencing of the CYP2B6 gene was performed in 12 and 35 selected individuals with high (S)-methadone plasma exposure and low (S)-methadone plasma exposure, respectively, from a previously described cohort of 276 patients undergoing methadone maintenance treatment. Methadone 141-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 23249875-2 2013 METHODS: Resequencing of the CYP2B6 gene was performed in 12 and 35 selected individuals with high (S)-methadone plasma exposure and low (S)-methadone plasma exposure, respectively, from a previously described cohort of 276 patients undergoing methadone maintenance treatment. Methadone 141-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 23249875-8 2013 CONCLUSION: Known genetic polymorphisms in CYP2B6 contribute toward explaining extreme (S)-methadone plasma levels observed in a cohort of patients following methadone maintenance treatment. Methadone 87-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 23249875-8 2013 CONCLUSION: Known genetic polymorphisms in CYP2B6 contribute toward explaining extreme (S)-methadone plasma levels observed in a cohort of patients following methadone maintenance treatment. Methadone 91-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 22830954-0 2013 Effects of clopidogrel and clarithromycin on the disposition of sibutramine and its active metabolites M1 and M2 in relation to CYP2B6*6 polymorphism. sibutramine 64-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-134 22830954-3 2013 Co-administration of clarithromycin, combined with CYP2B6*6/*6 genotype, led to highest concentration of sibutramine. sibutramine 105-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 22830954-5 2013 The CYP2B6*6/*6 subjects in the clopidogrel phase showed the highest molar AUC (M1 + M2) among three genotype groups throughout the three phases. Clopidogrel 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 22830954-6 2013 The exposure of sibutramine and its metabolites seemed to be associated with the CYP2B6 genotype. sibutramine 16-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 22830954-8 2013 These results suggest that the perturbation of CYP2B6 activity may contribute to the inter-individual variation of sibutramine drug responses although the clinical relevance is remained to be established. sibutramine 115-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 23088752-0 2013 Reactive intermediates produced from the metabolism of the vanilloid ring of capsaicinoids by p450 enzymes. Capsaicin 59-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-98 23088752-0 2013 Reactive intermediates produced from the metabolism of the vanilloid ring of capsaicinoids by p450 enzymes. Capsaicin 77-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-98 23088752-1 2013 This study characterized electrophilic and radical products derived from the metabolism of capsaicin by cytochrome P450 and peroxidase enzymes. Capsaicin 91-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-119 23088752-2 2013 Multiple glutathione and beta-mercaptoethanol conjugates (a.k.a., adducts), derived from the trapping of quinone methide and quinone intermediates of capsaicin, its analogue nonivamide, and O-demethylated and aromatic hydroxylated metabolites thereof, were produced by human liver microsomes and individual recombinant human P450 enzymes. Glutathione 9-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 325-329 23088752-2 2013 Multiple glutathione and beta-mercaptoethanol conjugates (a.k.a., adducts), derived from the trapping of quinone methide and quinone intermediates of capsaicin, its analogue nonivamide, and O-demethylated and aromatic hydroxylated metabolites thereof, were produced by human liver microsomes and individual recombinant human P450 enzymes. Mercaptoethanol 25-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 325-329 23088752-2 2013 Multiple glutathione and beta-mercaptoethanol conjugates (a.k.a., adducts), derived from the trapping of quinone methide and quinone intermediates of capsaicin, its analogue nonivamide, and O-demethylated and aromatic hydroxylated metabolites thereof, were produced by human liver microsomes and individual recombinant human P450 enzymes. quinone 105-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 325-329 23088752-2 2013 Multiple glutathione and beta-mercaptoethanol conjugates (a.k.a., adducts), derived from the trapping of quinone methide and quinone intermediates of capsaicin, its analogue nonivamide, and O-demethylated and aromatic hydroxylated metabolites thereof, were produced by human liver microsomes and individual recombinant human P450 enzymes. quinone 125-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 325-329 23088752-2 2013 Multiple glutathione and beta-mercaptoethanol conjugates (a.k.a., adducts), derived from the trapping of quinone methide and quinone intermediates of capsaicin, its analogue nonivamide, and O-demethylated and aromatic hydroxylated metabolites thereof, were produced by human liver microsomes and individual recombinant human P450 enzymes. Capsaicin 150-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 325-329 23088752-2 2013 Multiple glutathione and beta-mercaptoethanol conjugates (a.k.a., adducts), derived from the trapping of quinone methide and quinone intermediates of capsaicin, its analogue nonivamide, and O-demethylated and aromatic hydroxylated metabolites thereof, were produced by human liver microsomes and individual recombinant human P450 enzymes. nonivamide 174-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 325-329 23088752-7 2013 These data demonstrated that capsaicin and structurally similar analogues are converted to reactive intermediates by certain P450 enzymes, which may partially explain conflicting reports related to the cytotoxic, pro-carcinogenic, and chemoprotective effects of capsaicinoids in different cells and/or organ systems. Capsaicin 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-129 23088752-7 2013 These data demonstrated that capsaicin and structurally similar analogues are converted to reactive intermediates by certain P450 enzymes, which may partially explain conflicting reports related to the cytotoxic, pro-carcinogenic, and chemoprotective effects of capsaicinoids in different cells and/or organ systems. Capsaicin 262-275 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-129 23160467-1 2013 Cyclophosphamide (CPA) is one of the most widely used chemotherapeutic prodrugs that undergoes hepatic bioactivation mediated predominantly by cytochrome P450 (CYP) 2B6. Cyclophosphamide 0-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-168 23160467-1 2013 Cyclophosphamide (CPA) is one of the most widely used chemotherapeutic prodrugs that undergoes hepatic bioactivation mediated predominantly by cytochrome P450 (CYP) 2B6. Cyclophosphamide 18-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-168 23160467-2 2013 Given that the CYP2B6 gene is primarily regulated by the constitutive androstane receptor (CAR, NR1I3), we hypothesize that selective activation of CAR can enhance systemic exposure of the pharmacologically active 4-hydroxycyclophosamide (4-OH-CPA), with improved efficacy of CPA-based chemotherapy. 4-hydroxycyclophosamide 214-237 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 23193974-4 2013 Using purified P450, b(5), and reductase (POR) in reconstituted assays, the D58G/D65G double mutation, of residues located in a hydrophilic alpha-helix of b(5), totally abolished the ability to stimulate CYP2E1-catalyzed chlorzoxazone 6-hydroxylation. Chlorzoxazone 221-234 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-40 23160467-2 2013 Given that the CYP2B6 gene is primarily regulated by the constitutive androstane receptor (CAR, NR1I3), we hypothesize that selective activation of CAR can enhance systemic exposure of the pharmacologically active 4-hydroxycyclophosamide (4-OH-CPA), with improved efficacy of CPA-based chemotherapy. 4-oh-cpa 239-247 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 23160467-2 2013 Given that the CYP2B6 gene is primarily regulated by the constitutive androstane receptor (CAR, NR1I3), we hypothesize that selective activation of CAR can enhance systemic exposure of the pharmacologically active 4-hydroxycyclophosamide (4-OH-CPA), with improved efficacy of CPA-based chemotherapy. Cyclophosphamide 244-247 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 23268924-5 2013 In human liver microsomes, IC50 values of GTE were 5.9, 4.5, 48.7, 25.1 and 13.8 microg/mL, for CYP2B6, CYP2C8, CYP2C19, CYP2D6 and CYP3A, respectively. epigallocatechin gallate 42-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 23640958-0 2013 Efavirenz intoxication due to a new CYP2B6 constellation. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 22554354-7 2013 The 8,14-dihydroxyefavirenz : efavirenz AUC(0,120 h) ratio was significantly correlated with the weight-adjusted CL/F of efavirenz (r(2) 0.4, P < 0.05), differed with CYP2B6*6 genotype and was affected by clopidogrel pretreatment (P < 0.05) but not by itraconazole pretreatment. 8,14-Dihydroxyefavirenz 4-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-178 22554354-8 2013 CONCLUSIONS: The disposition of 8,14-dihydroxy-EFV appears to be sensitive to CYP2B6 activity alterations in human subjects. 8,14-dihydroxy-efv 32-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 22554354-9 2013 The 8,14-dihydroxyefaviremz : efavirenz AUC(0,120 h) ratio is attractive as a candidate phenotyping index for CYP2B6 activity in vivo. 8,14-dihydroxyefaviremz 4-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 23268924-2 2013 The objective of the present study was to evaluate the effects of green tea extract (GTE, total catechins 86.5%, w/w) and (-)-epigallocatechin-3-gallate (EGCG) on the activities of CYP2B6, CYP2C8, CYP2C19, CYP2D6 and CYP3A in vitro, using pooled human liver and intestinal microsomes. epigallocatechin gallate 85-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-187 23640947-6 2013 Among genes associated with metabolism of the most commonly applied anaesthetics such as propofol and sevoflurane, the following ones can be mentioned: CYP2E1, CYP2B6, CYP2C9, GSTP1, UGT1A9, SULT1A1 and NQO1. Propofol 89-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 160-166 23640947-6 2013 Among genes associated with metabolism of the most commonly applied anaesthetics such as propofol and sevoflurane, the following ones can be mentioned: CYP2E1, CYP2B6, CYP2C9, GSTP1, UGT1A9, SULT1A1 and NQO1. Sevoflurane 102-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 160-166 24455721-3 2013 The results demonstrated that the methadone maintenance dose, CYP2B6 785G allele, and ABCB1 2677T allele have positive effects on the methadone plasma concentration. Methadone 134-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 23257392-1 2013 The cytochrome P450 (P450, CYP) 2A6 inhibitor chalepensin was found to inhibit human CYP1A1, CYP1A2, CYP2A13, CYP2C9, CYP2D6, CYP2E1, and CYP3A4 to different extents. 3-(alpha,alpha-dimethylallyl)psoralen 46-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-35 23385314-2 2013 The present study showed that efavirenz was a potent competitive inhibitor of CYP2B6 (average K(i) = 1.68 microM in HLMs and K(i) = 1.38 microM in expressed CYP2B6) and CYP2C8 (K(i) = 4.78 microM in pooled HLMs and K(i) = 4.80 microM in HLMs with CYP2C8*3/*3 genotype). efavirenz 30-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 23385314-2 2013 The present study showed that efavirenz was a potent competitive inhibitor of CYP2B6 (average K(i) = 1.68 microM in HLMs and K(i) = 1.38 microM in expressed CYP2B6) and CYP2C8 (K(i) = 4.78 microM in pooled HLMs and K(i) = 4.80 microM in HLMs with CYP2C8*3/*3 genotype). efavirenz 30-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 23268924-2 2013 The objective of the present study was to evaluate the effects of green tea extract (GTE, total catechins 86.5%, w/w) and (-)-epigallocatechin-3-gallate (EGCG) on the activities of CYP2B6, CYP2C8, CYP2C19, CYP2D6 and CYP3A in vitro, using pooled human liver and intestinal microsomes. Catechin 96-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-187 23268924-2 2013 The objective of the present study was to evaluate the effects of green tea extract (GTE, total catechins 86.5%, w/w) and (-)-epigallocatechin-3-gallate (EGCG) on the activities of CYP2B6, CYP2C8, CYP2C19, CYP2D6 and CYP3A in vitro, using pooled human liver and intestinal microsomes. epigallocatechin gallate 122-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-187 23268924-9 2013 These results suggest that green tea catechins cause clinically relevant interactions with substrates for CYP2B6 and CYP2C8 in addition to CYP3A. Catechin 37-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 23268924-2 2013 The objective of the present study was to evaluate the effects of green tea extract (GTE, total catechins 86.5%, w/w) and (-)-epigallocatechin-3-gallate (EGCG) on the activities of CYP2B6, CYP2C8, CYP2C19, CYP2D6 and CYP3A in vitro, using pooled human liver and intestinal microsomes. epigallocatechin gallate 154-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-187 23149928-5 2012 Genetic variation in CYP2B6, the enzyme that metabolizes bupropion to hydroxybupropion, was identified as a significant source of variability in hydroxybupropion formation. Bupropion 57-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 23648676-4 2013 In the present study, P450 metabolic activity was investigated in the small intestine of MacfaCAM and MacfaCHN, and cynomolgus macaques bred in Indonesia (MacfaIDN) using P450 substrates, including 7-ethoxyresorufin, coumarin, bupropion, paclitaxel, diclofenac, S-mephenytoin, bufuralol, chlorzoxazone, and testosterone. Paclitaxel 238-248 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 23648676-4 2013 In the present study, P450 metabolic activity was investigated in the small intestine of MacfaCAM and MacfaCHN, and cynomolgus macaques bred in Indonesia (MacfaIDN) using P450 substrates, including 7-ethoxyresorufin, coumarin, bupropion, paclitaxel, diclofenac, S-mephenytoin, bufuralol, chlorzoxazone, and testosterone. Diclofenac 250-260 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 23648676-4 2013 In the present study, P450 metabolic activity was investigated in the small intestine of MacfaCAM and MacfaCHN, and cynomolgus macaques bred in Indonesia (MacfaIDN) using P450 substrates, including 7-ethoxyresorufin, coumarin, bupropion, paclitaxel, diclofenac, S-mephenytoin, bufuralol, chlorzoxazone, and testosterone. Mephenytoin 262-275 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 23648676-4 2013 In the present study, P450 metabolic activity was investigated in the small intestine of MacfaCAM and MacfaCHN, and cynomolgus macaques bred in Indonesia (MacfaIDN) using P450 substrates, including 7-ethoxyresorufin, coumarin, bupropion, paclitaxel, diclofenac, S-mephenytoin, bufuralol, chlorzoxazone, and testosterone. bufuralol 277-286 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 23648676-4 2013 In the present study, P450 metabolic activity was investigated in the small intestine of MacfaCAM and MacfaCHN, and cynomolgus macaques bred in Indonesia (MacfaIDN) using P450 substrates, including 7-ethoxyresorufin, coumarin, bupropion, paclitaxel, diclofenac, S-mephenytoin, bufuralol, chlorzoxazone, and testosterone. Chlorzoxazone 288-301 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 23648676-4 2013 In the present study, P450 metabolic activity was investigated in the small intestine of MacfaCAM and MacfaCHN, and cynomolgus macaques bred in Indonesia (MacfaIDN) using P450 substrates, including 7-ethoxyresorufin, coumarin, bupropion, paclitaxel, diclofenac, S-mephenytoin, bufuralol, chlorzoxazone, and testosterone. Testosterone 307-319 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 22519658-1 2012 AIMS: This study aimed to investigate the effect of metamizole on bupropion hydroxylation related to different CYP2B6 genotype groups in healthy volunteers. Dipyrone 52-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 22519658-1 2012 AIMS: This study aimed to investigate the effect of metamizole on bupropion hydroxylation related to different CYP2B6 genotype groups in healthy volunteers. Bupropion 66-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 22519658-10 2012 CONCLUSIONS: Oral administration of metamizole for 4 days significantly altered the pharmacokinetics of both bupropion and its active metabolite, 4-hydroxybupropion, and significantly increased the CYP2B6-catalyzed bupropion hydroxylation in all of the subjects. Dipyrone 36-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 23648676-4 2013 In the present study, P450 metabolic activity was investigated in the small intestine of MacfaCAM and MacfaCHN, and cynomolgus macaques bred in Indonesia (MacfaIDN) using P450 substrates, including 7-ethoxyresorufin, coumarin, bupropion, paclitaxel, diclofenac, S-mephenytoin, bufuralol, chlorzoxazone, and testosterone. ethoxyresorufin 198-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 23648676-4 2013 In the present study, P450 metabolic activity was investigated in the small intestine of MacfaCAM and MacfaCHN, and cynomolgus macaques bred in Indonesia (MacfaIDN) using P450 substrates, including 7-ethoxyresorufin, coumarin, bupropion, paclitaxel, diclofenac, S-mephenytoin, bufuralol, chlorzoxazone, and testosterone. coumarin 217-225 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 23648676-4 2013 In the present study, P450 metabolic activity was investigated in the small intestine of MacfaCAM and MacfaCHN, and cynomolgus macaques bred in Indonesia (MacfaIDN) using P450 substrates, including 7-ethoxyresorufin, coumarin, bupropion, paclitaxel, diclofenac, S-mephenytoin, bufuralol, chlorzoxazone, and testosterone. Bupropion 227-236 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 23665929-7 2013 Furthermore, the hCAR-mediated basal induction of endogenous cytochrome P450 2B6 (CYP2B6) mRNA was adversely affected by co-treatment with TO901317. T0901317 139-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-80 23665929-7 2013 Furthermore, the hCAR-mediated basal induction of endogenous cytochrome P450 2B6 (CYP2B6) mRNA was adversely affected by co-treatment with TO901317. T0901317 139-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 22926004-9 2013 Women with the CYP2B6 Q172 variant GT genotype have consistently higher L/D values for S-methadone across both pregnancy and postpartum. Methadone 87-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 22950382-7 2012 The genotype CYP2B6-516 guanine/guanine (G/G), guanine/thymidine (G/T), and T/T were found in 49%, 37%, and 14% of patients, respectively. Guanine 24-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 22950382-7 2012 The genotype CYP2B6-516 guanine/guanine (G/G), guanine/thymidine (G/T), and T/T were found in 49%, 37%, and 14% of patients, respectively. Guanine 32-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 22950382-7 2012 The genotype CYP2B6-516 guanine/guanine (G/G), guanine/thymidine (G/T), and T/T were found in 49%, 37%, and 14% of patients, respectively. Guanine 32-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 22950382-11 2012 CYP2B6-516G > T polymorphism was the only factor associated with high plasma EFV levels. efavirenz 80-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22936314-0 2012 Inhibition of bupropion metabolism by selegiline: mechanism-based inactivation of human CYP2B6 and characterization of glutathione and peptide adducts. Selegiline 38-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 22936314-2 2012 Bupropion, an antidepressant, often used to treat patients in conjunction with selegiline, is metabolized primarily by CYP2B6. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 22936314-2 2012 Bupropion, an antidepressant, often used to treat patients in conjunction with selegiline, is metabolized primarily by CYP2B6. Selegiline 79-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 22936314-3 2012 The effect of selegiline on the enzymatic activity of human cytochrome CYP2B6 in a reconstituted system and its effect on the metabolism of bupropion were examined. Selegiline 14-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 22936314-4 2012 Selegiline was found to be a mechanism-based inactivator of the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation (7-EFC) activity of CYP2B6 as well as bupropion metabolism. Selegiline 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 22936314-4 2012 Selegiline was found to be a mechanism-based inactivator of the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation (7-EFC) activity of CYP2B6 as well as bupropion metabolism. 7-ethoxy-4-trifluoromethylcoumarin 64-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 22936314-6 2012 In standard inhibition assays, selegiline increased the K(m) of CYP2B6 for bupropion from 10 to 92 muM and decreased the k(cat) by ~50%. Selegiline 31-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 22936314-6 2012 In standard inhibition assays, selegiline increased the K(m) of CYP2B6 for bupropion from 10 to 92 muM and decreased the k(cat) by ~50%. Bupropion 75-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 22942317-6 2012 Earlier studies from our laboratory have shown that tamoxifen is a mechanism-based inactivator of CYP2B6. Tamoxifen 52-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 22942317-9 2012 The metabolic activation of tamoxifen in the CYP2B6 reconstituted system also resulted in the formation of an adduct to the P4502B6 apoprotein, which was identified using liquid chromatography mass spectrometry. Tamoxifen 28-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 23149928-5 2012 Genetic variation in CYP2B6, the enzyme that metabolizes bupropion to hydroxybupropion, was identified as a significant source of variability in hydroxybupropion formation. hydroxybupropion 70-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 23149928-5 2012 Genetic variation in CYP2B6, the enzyme that metabolizes bupropion to hydroxybupropion, was identified as a significant source of variability in hydroxybupropion formation. hydroxybupropion 145-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 23149928-6 2012 Our data indicate that hydroxybupropion contributes to the pharmacologic effects of bupropion for smoking cessation, and that variability in response to bupropion treatment is related to variability in CYP2B6-mediated hydroxybupropion formation. hydroxybupropion 218-234 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 202-208 22775490-0 2012 Bioactivation of chlorpyrifos by CYP2B6 variants. Chlorpyrifos 17-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 22775490-3 2012 CYP2B6.4,.5,.7, and .18 were over-expressed in mammalian COS-1 cells to assess the impact of CYP2B6 variants on the K(m) and V(max) for bioactivation of CPF. carbonyl sulfide 57-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22775490-10 2012 Variants of CYP2B6 have altered capacity to bioactivate CPF and may affect individual susceptibility by altering the V(max) for CPF-O formation. cpf-o 128-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 23129539-5 2012 Although type II ligands typically block P450 activity, we report here that type II ligation can be harnessed to achieve just the opposite, that is, to favor biocatalysis and afford predictable oxidation of small hydrocarbon substrates with P450 2E1. Hydrocarbons 213-224 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 23129539-5 2012 Although type II ligands typically block P450 activity, we report here that type II ligation can be harnessed to achieve just the opposite, that is, to favor biocatalysis and afford predictable oxidation of small hydrocarbon substrates with P450 2E1. Hydrocarbons 213-224 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 241-245 22671777-4 2012 Concentrations of PQ were measured after incubation with both human liver microsomes (HLMs) and expressed cytochrome P450 enzymes (P450s). piperaquine 18-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-121 22859722-4 2012 Every P450 tested except CYP2E1 was capable of raloxifene bioactivation, based on glutathione adduct formation. Raloxifene Hydrochloride 47-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-10 22859722-4 2012 Every P450 tested except CYP2E1 was capable of raloxifene bioactivation, based on glutathione adduct formation. Glutathione 82-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-10 22859722-8 2012 For each P450 enzyme, proposed substrate/metabolite access channels were mapped and active site cysteines were identified, which revealed that only CYP2C8 and CYP3A4 possess accessible cysteine residues near the active site cavities, a result consistent with the observed kinetics. Cysteine 96-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-13 22859722-8 2012 For each P450 enzyme, proposed substrate/metabolite access channels were mapped and active site cysteines were identified, which revealed that only CYP2C8 and CYP3A4 possess accessible cysteine residues near the active site cavities, a result consistent with the observed kinetics. Cysteine 96-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-13 23104099-9 2012 In bivariate linear regression, CYP2B6 516G T (CYP2B6*6) was associated with lower nevirapine clearances (P=3.5x10). Nevirapine 83-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 23104099-9 2012 In bivariate linear regression, CYP2B6 516G T (CYP2B6*6) was associated with lower nevirapine clearances (P=3.5x10). Nevirapine 83-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 22851615-5 2012 The majority of the analyzed genes was either absent or expressed at low levels in the THLE-null and THLE-P450 cells, apart from housekeeping genes and the individual transfected P450s. thle 101-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-110 22859722-0 2012 Cytochrome p450 architecture and cysteine nucleophile placement impact raloxifene-mediated mechanism-based inactivation. Raloxifene Hydrochloride 71-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 22859722-3 2012 Raloxifene, a drug known to undergo CYP3A-mediated reactive metabolite formation and time-dependent inhibition in vitro, was used to explore the potential for bioactivation and enzyme inactivation of additional P450 enzymes (CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A5). Raloxifene Hydrochloride 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 211-215 22671777-6 2012 Miconazole, at nonspecific P450 inhibitory concentrations, resulted in almost complete inhibition of PQ metabolism. Miconazole 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 22671777-8 2012 Using a mixture of recombinant P450 enzymes, turnover for PQ metabolism was estimated as 0.0099 min(-1); recombinant CYP3A4 had a higher metabolic rate (0.017 min(-1)) than recombinant CYP2C8 (p < .0001). piperaquine 58-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 22926595-10 2012 In patients presenting the CYP2B6 *6/*6 genotype (n = 8), S-OH-MIR concentrations were higher those in the other patients (n = 37). s-oh-mir 58-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 23961363-3 2012 The prevalence of poor and rapid metabolizers was determined in the target population for the category of drugs metabolized by CYP2B6 by measuring plasma bupropion, a drug metabolized by CYP2B6, and its metabolite. Bupropion 154-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 23961363-5 2012 CYP2B6 activity was measured as hydroxybupropion/bupropion ratio, and volunteers were categorized as rapid or poor metabolizers on the basis of cutoff value of log (hydroxybupropion/bupropion). hydroxybupropion 32-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23961363-5 2012 CYP2B6 activity was measured as hydroxybupropion/bupropion ratio, and volunteers were categorized as rapid or poor metabolizers on the basis of cutoff value of log (hydroxybupropion/bupropion). Bupropion 39-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23961363-5 2012 CYP2B6 activity was measured as hydroxybupropion/bupropion ratio, and volunteers were categorized as rapid or poor metabolizers on the basis of cutoff value of log (hydroxybupropion/bupropion). Bupropion 49-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23961363-8 2012 Cutoff value defined in this study can be used as a tool for evaluating the status of CYP2B6 using bupropion as a probe drug. Bupropion 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 22752007-3 2012 Our previous study clarified the sequential metabolism of sesamin by cytochrome P450 (P450) and UDP-glucuronosyltransferase in human liver. sesamin 58-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-91 23298916-2 2012 Their multiethnic populations suggest complexity due to the genetic polymorphism, such as CYP2B6 that metabolizes methadone and anti-retroviral. Methadone 114-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 23298916-3 2012 AIMS: Our aim was to explore the genetic polymorphism of CYP2B6 among Malays, Chinese, Indians, and opiate-dependent individuals in Malaysia. Opiate Alkaloids 100-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 23298916-11 2012 Reduced activity CYP2B6*6 occurred in 13% to 26% among Malays, Chinese, Indians and opiate-dependent individuals. Opiate Alkaloids 84-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 23298916-13 2012 CONCLUSIONS: The relative commonness of reduced-activity CYP2B6 alleles in our study called for attention in terms of dosage requirements for MMT and ARV in Malaysia. omega-N-Allylarginine 150-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 22738989-12 2012 Collectively, the results show that the oxidative metabolism of BDE-99 by human liver microsomes is catalyzed solely by CYP2B6 and is an important determinant of the toxicity and bioaccumulation of BDE-99 in humans. 2,2',4,4',5-brominated diphenyl ether 64-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 22738989-12 2012 Collectively, the results show that the oxidative metabolism of BDE-99 by human liver microsomes is catalyzed solely by CYP2B6 and is an important determinant of the toxicity and bioaccumulation of BDE-99 in humans. 2,2',4,4',5-brominated diphenyl ether 198-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 22909231-0 2012 Conformational adaptation of human cytochrome P450 2B6 and rabbit cytochrome P450 2B4 revealed upon binding multiple amlodipine molecules. Amlodipine 117-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-54 22738989-0 2012 Oxidative metabolism of BDE-99 by human liver microsomes: predominant role of CYP2B6. 2,2',4,4',5-brominated diphenyl ether 24-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 22909231-1 2012 Structures of human cytochrome P450 2B6 and rabbit cytochrome P450 2B4 in complex with two molecules of the calcium channel blocker amlodipine have been determined by X-ray crystallography. Amlodipine 132-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-39 22380717-12 2012 CYP2B6*9 variants vs. wildtype were found to have decreased elimination rate constant (P = 0.0005, 95% CI 0.033, 0.103), increased V(d) (P = 0.0271, 95% CI -57.5, -4.2) and decreased C(max) (P = 0.0176, 95% CI 0.696, 6179) for cyclophosphamide. Cyclophosphamide 227-243 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22910411-3 2012 Using HepG2 cells expressing human CAR in the presence of tetracycline, we showed that knockdown of DP97 with small interfering RNAs suppressed tetracycline-inducible mRNA expression of CYP2B6 and UGT1A1 but not CYP3A4. Tetracycline 144-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 186-192 22380717-17 2012 The CYP2B6*9 and ABCB1 C3435T polymorphisms alter the pharmacokinetics of cyclophosphamide and 4-hydroxycyclophosphamide in glomerulonephritis. Cyclophosphamide 74-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 22380717-17 2012 The CYP2B6*9 and ABCB1 C3435T polymorphisms alter the pharmacokinetics of cyclophosphamide and 4-hydroxycyclophosphamide in glomerulonephritis. 4-hydroxycyclophosphamide 95-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 22679214-4 2012 To better understand the autoinduction and metabolic drug-drug interactions (DDIs), we evaluated the P450s (particularly CYP2B6 and CYP3A4) inhibited or induced by two artemisinin drugs, Qing-hao-su (QHS) and dihydroartemisinin (DHA) using human liver microsome, recombinant P450 enzymes, and primary human hepatocytes. artemisinin 168-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-127 22685215-0 2012 Mechanism-based inactivation of cytochrome P450 2B6 by methadone through destruction of prosthetic heme. Methadone 55-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-51 22685215-0 2012 Mechanism-based inactivation of cytochrome P450 2B6 by methadone through destruction of prosthetic heme. Heme 99-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-51 22685215-4 2012 However, recent clinical data have indicated that CYP2B6 is actually the major isoform responsible for methadone metabolism and clearance in vivo. Methadone 103-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 22685215-5 2012 In this study, methadone was shown to act as a mechanism-based inactivator of CYP2B6. Methadone 15-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 22685215-6 2012 Methadone inactivates CYP2B6 in a time-, concentration-, and NADPH-dependent manner with a K(I) = 10.0 muM and k(inact) = 0.027 min-1. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 22685215-6 2012 Methadone inactivates CYP2B6 in a time-, concentration-, and NADPH-dependent manner with a K(I) = 10.0 muM and k(inact) = 0.027 min-1. NADP 61-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 22685215-7 2012 The loss of CYP2B6 activity in the presence of methadone and NADPH occurred with concomitant loss of the reduced CO spectrum of the P450. Methadone 47-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 22685215-7 2012 The loss of CYP2B6 activity in the presence of methadone and NADPH occurred with concomitant loss of the reduced CO spectrum of the P450. NADP 61-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 22685215-9 2012 High-performance liquid chromatography analysis of the native heme of the inactivated CYP2B6 demonstrated that approximately 75% loss of heme was accompanied by comparable inactivation of CYP2B6. Heme 62-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 22685215-9 2012 High-performance liquid chromatography analysis of the native heme of the inactivated CYP2B6 demonstrated that approximately 75% loss of heme was accompanied by comparable inactivation of CYP2B6. Heme 137-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 22685215-11 2012 The evidence strongly suggests that destruction of prosthetic heme is the underlying mechanism leading to the inactivation of CYP2B6 by methadone. Heme 62-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 22685215-11 2012 The evidence strongly suggests that destruction of prosthetic heme is the underlying mechanism leading to the inactivation of CYP2B6 by methadone. Methadone 136-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 22679214-4 2012 To better understand the autoinduction and metabolic drug-drug interactions (DDIs), we evaluated the P450s (particularly CYP2B6 and CYP3A4) inhibited or induced by two artemisinin drugs, Qing-hao-su (QHS) and dihydroartemisinin (DHA) using human liver microsome, recombinant P450 enzymes, and primary human hepatocytes. artemisinin 168-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 22428615-11 2012 Moreover, incubation with inhibitors of CYP3A, CYP2B6 and CYP2C19 significantly reduced clopidogrel bioactivation while a PON1 inhibitor, EDTA, had only a weak inhibitory effect. Clopidogrel 88-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 23439719-6 2012 Associations were identified with HLA-Cw*04, HLA-B*35, HLA-DRB*01 and CYP2B6 516G>T (rs3745274); however, positive predictive values for these genetic markers were low, and most nevirapine-associated adverse events occurred in patients without these markers. Nevirapine 181-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 22428615-13 2012 Moreover, CYP2C19, CYP2B6 and CYP3A play important roles in the bioactivation of clopidogrel. Clopidogrel 81-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 22484313-0 2012 Estradiol induces cytochrome P450 2B6 expression at high concentrations: implication in estrogen-mediated gene regulation in pregnancy. Estradiol 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-37 22584220-1 2012 We reported previously the formation of a glutathionyl conjugate of the active metabolite (AM) of clopidogrel and the covalent modification of a cysteinyl residue of human cytochrome P450 2B6 in a reconstituted system (Mol Pharmacol 80:839-847, 2011). lysyl-cysteinyl-cysteinyl-arginyl-cysteinyl-lysine 145-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-191 22722506-3 2012 The complete metabolic disposition of methadone is likely to involve a number of enzymes, including specifically CYP2B6. Methadone 38-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 22484313-2 2012 We have found that 17beta-estradiol (E2) upregulates expression of the major drug-metabolizing enzyme CYP2B6 in primary human hepatocytes. Estradiol 19-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 22484313-8 2012 Concurrent activation of both ER and CAR by E2 enhanced CYP2B6 expression in a synergistic manner. Estradiol 44-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 22531045-7 2012 Carnosic acid also induced CYP2B6 and CYP3A4 mRNA and enzyme activity in a dose-dependent manner. salvin 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 22680342-0 2012 Influence of CYP2B6 and ABCB1 SNPs on nevirapine plasma concentrations in Burundese HIV-positive patients using dried sample spot devices. Nevirapine 38-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 22680342-1 2012 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: Nevirapine pharmacokinetics are affected by several factors including CYP2B6 single nucleotide polymorphisms (SNPs). Nevirapine 44-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 22680342-7 2012 CYP2B6 (both at position 516 and 983) but not ABCB1 (3435 and 1236) SNPs as well as age correlate with higher nevirapine exposure. Nevirapine 110-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22233204-8 2012 METHODS: In vitro experiments were conducted to assess the inhibition and induction potential of tofacitinib for major drug metabolizing enzymes (CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and CYP3A4). tofacitinib 97-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-160 22531045-8 2012 At 10 muM, carnosic acid increased CYP2B6 enzyme activity 61.6 and 49.3% in two donors compared with phenobarbital, and it increased CYP3A enzyme activity 82.6 and 142% compared with rifampicin. salvin 11-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 22450566-3 2012 The objective of the present work was to evaluate the impact of vanillin and ethyl vanillin on the activities of CYP2C9, CYP2E1, CYP3A4, CYP2B6 and CYP1A2 in human liver microsomes (HLM) in vitro, and impact on the activities of CYP1A2, CYP2C, CYP3A and CYP2E1 in rat liver microsomes (RLM) in vivo. vanillin 64-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 22568496-0 2012 Microarray of human P450 with an integrated oxygen sensing film for high-throughput detection of metabolic activities. Oxygen 44-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 22568496-6 2012 By normalizing the responses with the amounts of oxygen sensor and P450 enzymes in microwells, we could obtain fluorescence enhancement patterns that were characteristic to the combination of P450 isoforms and substrate material. Oxygen 49-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-196 22313038-0 2012 Different effects of proton pump inhibitors and famotidine on the clopidogrel metabolic activation by recombinant CYP2B6, CYP2C19 and CYP3A4. Famotidine 48-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 22313038-0 2012 Different effects of proton pump inhibitors and famotidine on the clopidogrel metabolic activation by recombinant CYP2B6, CYP2C19 and CYP3A4. Clopidogrel 66-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 22313038-1 2012 Inhibitory potential of proton pump inhibitors (PPIs) and famotidine, an H(2) receptor antagonist, on the metabolic activation of clopidogrel was evaluated using recombinant CYP2B6, CYP2C19 and CYP3A4. Clopidogrel 130-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 174-180 22313038-3 2012 Omeprazole potently inhibited clopidogrel activation by CYP2C19 with an IC(50) of 12.8 mumol/L and more weakly inhibited that by CYP2B6 and CYP3A4. Omeprazole 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 22450566-3 2012 The objective of the present work was to evaluate the impact of vanillin and ethyl vanillin on the activities of CYP2C9, CYP2E1, CYP3A4, CYP2B6 and CYP1A2 in human liver microsomes (HLM) in vitro, and impact on the activities of CYP1A2, CYP2C, CYP3A and CYP2E1 in rat liver microsomes (RLM) in vivo. ethyl vanillin 77-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 21814747-4 2012 The selectivity of the most potent aromatase inhibitor identified, norendoxifen, was characterized by studying its ability to inhibit CYP450 enzymes important in clinical drug-drug interactions, including CYP2B6, 2C9, 2C19, 2D6, and 3A. N,N-didesmethyl-4-hydroxytamoxifen 67-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 205-211 22569203-0 2012 CYP2A6 and CYP2B6 genetic variation and its association with nicotine metabolism in South Western Alaska Native people. Nicotine 61-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 22569203-2 2012 Variations in the CYP2A6 and CYP2B6 genes, encoding enzymes responsible for nicotine metabolic inactivation and procarcinogen activation, have not been characterized in AN and may contribute toward the increased risk. Nicotine 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 22569203-6 2012 Nicotine metabolism [as measured by the plasma and urinary ratio of metabolites trans-3"-hydroxycotinine to cotinine (3HC/COT)] was significantly associated with CYP2A6 (P<0.001), but not CYP2B6 genotype (P=0.95) when controlling for known covariates. Nicotine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 191-197 22339467-2 2012 CYP2B6 mediates oxidation of bupropion to hydroxybupropion, but the enzyme(s) catalyzing carbonyl reduction of bupropion to erythro- and threohydrobupropion in human liver is unknown. Bupropion 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22339467-2 2012 CYP2B6 mediates oxidation of bupropion to hydroxybupropion, but the enzyme(s) catalyzing carbonyl reduction of bupropion to erythro- and threohydrobupropion in human liver is unknown. hydroxybupropion 42-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22339467-2 2012 CYP2B6 mediates oxidation of bupropion to hydroxybupropion, but the enzyme(s) catalyzing carbonyl reduction of bupropion to erythro- and threohydrobupropion in human liver is unknown. Bupropion 49-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22339467-2 2012 CYP2B6 mediates oxidation of bupropion to hydroxybupropion, but the enzyme(s) catalyzing carbonyl reduction of bupropion to erythro- and threohydrobupropion in human liver is unknown. erythro- and threohydrobupropion 124-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22349139-8 2012 Inhibitor screening kits were used to quantify the inhibition of CYP3A4, CYP2C19, CYP2B6 and CYP2D6 by bisnortilidine. bisnortilidine 103-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 22349139-11 2012 Assays with recombinant CYPs verified that the N-demethylation is catalysed by CYP3A4, CYP2C19 and CYP2B6. Nitrogen 47-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 22349139-12 2012 Our results also demonstrated that the metabolism from tilidine to nortilidine is not only mediated by CYP3A4 and CYP2C19, but also by CYP2B6. Tilidine 55-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 22349139-12 2012 Our results also demonstrated that the metabolism from tilidine to nortilidine is not only mediated by CYP3A4 and CYP2C19, but also by CYP2B6. nortilidine 67-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 22349139-13 2012 Moreover, bisnortilidine is a weak inhibitor of CYP3A4 and CYP2B6, a strong inhibitor of CYP2D6, but not an inhibitor of CYP2C19. bisnortilidine 10-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 22390216-1 2012 The antineoplastic alkaloid ellipticine is a prodrug, whose pharmacological efficiency is dependent on its cytochrome P450 (P450)- and/or peroxidase-mediated activation in target tissues. Alkaloids 19-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-148 22390216-1 2012 The antineoplastic alkaloid ellipticine is a prodrug, whose pharmacological efficiency is dependent on its cytochrome P450 (P450)- and/or peroxidase-mediated activation in target tissues. ellipticine 28-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-148 22232427-7 2012 Inhibition of efavirenz 8-hydroxylation by voriconazole was significantly greater in HLMs with the CYP2B6*6 allele (K(i) = 1.6 +- 0.8 muM) than HLMs with CYP2B6*1/*1 genotype (K(i) = 3.0 +- 1.1 muM). Voriconazole 43-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-160 22511698-2 2012 Methadone is primarily metabolized by N-demethylation to an inactive metabolite 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidene (EDDP) by CYP3A4 and CYP2B6. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 22511698-2 2012 Methadone is primarily metabolized by N-demethylation to an inactive metabolite 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidene (EDDP) by CYP3A4 and CYP2B6. Nitrogen 38-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 22511698-2 2012 Methadone is primarily metabolized by N-demethylation to an inactive metabolite 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidene (EDDP) by CYP3A4 and CYP2B6. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidene 80-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 22511698-2 2012 Methadone is primarily metabolized by N-demethylation to an inactive metabolite 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidene (EDDP) by CYP3A4 and CYP2B6. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 131-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 22398970-8 2012 Efavirenz induced hepatocyte CYP2B6 and CYP3A4 expression, activity, and methadone N-demethylation. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 22398970-10 2012 Methadone disposition was most consistent with efavirenz induction of hepatic CYP2B6-mediated methadone N-demethylation. Methadone 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 22398970-10 2012 Methadone disposition was most consistent with efavirenz induction of hepatic CYP2B6-mediated methadone N-demethylation. Methadone 94-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 22266842-2 2012 The oxidative metabolism of lorcaserin, mediated by recombinant human cytochrome P450 (P450) and flavin-containing monooxygenase (FMO) enzymes, was examined in vitro to identify the enzymes involved in the generation of its primary oxidative metabolites, N-hydroxylorcaserin, 7-hydroxylorcaserin, 5-hydroxylorcaserin, and 1-hydroxylorcaserin. lorcaserin 28-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-92 22245954-1 2012 PURPOSE: The prodrug cyclophosphamide is metabolized by cytochrome P450(CYP)2B6 to the active metabolite, 4-hydroxycyclophosphamide (4-OH), and by CYP3A4/5 to toxic chloracetaldehyde and 2-dechloroethylcyclophosphamide (DCE). Cyclophosphamide 21-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-79 22245954-1 2012 PURPOSE: The prodrug cyclophosphamide is metabolized by cytochrome P450(CYP)2B6 to the active metabolite, 4-hydroxycyclophosphamide (4-OH), and by CYP3A4/5 to toxic chloracetaldehyde and 2-dechloroethylcyclophosphamide (DCE). 4-hydroxycyclophosphamide 106-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-79 22245954-1 2012 PURPOSE: The prodrug cyclophosphamide is metabolized by cytochrome P450(CYP)2B6 to the active metabolite, 4-hydroxycyclophosphamide (4-OH), and by CYP3A4/5 to toxic chloracetaldehyde and 2-dechloroethylcyclophosphamide (DCE). 4-hydroxycyclophosphamide 133-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-79 22245954-1 2012 PURPOSE: The prodrug cyclophosphamide is metabolized by cytochrome P450(CYP)2B6 to the active metabolite, 4-hydroxycyclophosphamide (4-OH), and by CYP3A4/5 to toxic chloracetaldehyde and 2-dechloroethylcyclophosphamide (DCE). chloroacetaldehyde 165-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-79 22245954-1 2012 PURPOSE: The prodrug cyclophosphamide is metabolized by cytochrome P450(CYP)2B6 to the active metabolite, 4-hydroxycyclophosphamide (4-OH), and by CYP3A4/5 to toxic chloracetaldehyde and 2-dechloroethylcyclophosphamide (DCE). 2-dechloroethylcyclophosphamide 187-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-79 22245954-1 2012 PURPOSE: The prodrug cyclophosphamide is metabolized by cytochrome P450(CYP)2B6 to the active metabolite, 4-hydroxycyclophosphamide (4-OH), and by CYP3A4/5 to toxic chloracetaldehyde and 2-dechloroethylcyclophosphamide (DCE). 2-dechloroethylcyclophosphamide 220-223 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-79 22232427-4 2012 Significantly higher V(max) and K(m) values for 8-hydroxyefavirenz formation and ~2-fold lower intrinsic clearance (Cl(int)) were noted in expressed CYP2B6.6 protein (-b5) compared with that of CYP2B6.1 protein (-b5); this effect was abolished by Cyt b5. 8-hydroxyefavirenz 48-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 22232427-5 2012 The V(max) and Cl(int) values for 4-hydroxybupropion formation were significantly higher in CYP2B6.6 than in CYP2B6.1 protein, with no difference in K(m), whereas coexpression with Cyt b5 reversed the genetic effect on these kinetic parameters. hydroxybupropion 34-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 22232427-5 2012 The V(max) and Cl(int) values for 4-hydroxybupropion formation were significantly higher in CYP2B6.6 than in CYP2B6.1 protein, with no difference in K(m), whereas coexpression with Cyt b5 reversed the genetic effect on these kinetic parameters. hydroxybupropion 34-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 22232427-6 2012 In HLMs, CYP2B6*6/*6 genotype was associated with markedly lower V(max) (and moderate increase in K(m)) and thus lower Cl(int) values for efavirenz and bupropion metabolism, but no difference in catalytic properties was noted between CYP2B6*1/*1 and CYP2B6*1/*6 genotypes. Bupropion 152-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 22232427-7 2012 Inhibition of efavirenz 8-hydroxylation by voriconazole was significantly greater in HLMs with the CYP2B6*6 allele (K(i) = 1.6 +- 0.8 muM) than HLMs with CYP2B6*1/*1 genotype (K(i) = 3.0 +- 1.1 muM). Voriconazole 43-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 22266842-4 2012 In 16 individual human liver microsomal preparations (HLM), formation of N-hydroxylorcaserin was correlated with CYP2B6, 7-hydroxylorcaserin was correlated with CYP2D6, 5-hydroxylorcaserin was correlated with CYP1A2 and CYP3A4, and 1-hydroxylorcaserin was correlated with CYP3A4 activity at 10.0 muM lorcaserin. N-Hydroxy Lorcaserin 73-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 22266842-4 2012 In 16 individual human liver microsomal preparations (HLM), formation of N-hydroxylorcaserin was correlated with CYP2B6, 7-hydroxylorcaserin was correlated with CYP2D6, 5-hydroxylorcaserin was correlated with CYP1A2 and CYP3A4, and 1-hydroxylorcaserin was correlated with CYP3A4 activity at 10.0 muM lorcaserin. lorcaserin 82-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 22266842-10 2012 Multiple human P450 and FMO enzymes catalyze the formation of four primary oxidative metabolites of lorcaserin, suggesting that lorcaserin has a low probability of drug-drug interactions by concomitant medications. lorcaserin 100-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-27 22266842-10 2012 Multiple human P450 and FMO enzymes catalyze the formation of four primary oxidative metabolites of lorcaserin, suggesting that lorcaserin has a low probability of drug-drug interactions by concomitant medications. lorcaserin 128-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-27 22281205-0 2012 Effect of CYP2B6*6 and CYP2C19*2 genotype on chlorpyrifos metabolism. Chlorpyrifos 45-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 22269145-0 2012 Biotransformation of the antiretroviral drug etravirine: metabolite identification, reaction phenotyping, and characterization of autoinduction of cytochrome P450-dependent metabolism. etravirine 45-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-162 22269145-3 2012 Incubations with cDNA-expressed P450 isozymes and chemical inhibition studies using HLMs indicated that CYP2C19 is primarily responsible for the formation of both the major monohydroxylated and dihydroxylated metabolites of ETR. etravirine 224-227 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 22310298-9 2012 CYP2B6 was induced fairly equally by organophosphate, carbamate and pyrethroid compounds. Organophosphates 37-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22310298-9 2012 CYP2B6 was induced fairly equally by organophosphate, carbamate and pyrethroid compounds. Carbamates 54-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22310298-9 2012 CYP2B6 was induced fairly equally by organophosphate, carbamate and pyrethroid compounds. Pyrethrins 68-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 22310298-11 2012 The urea herbicide diuron and the triazine herbicide atrazine induced CYP2B6 mRNA more than 10-fold, but did not activate CAR indicating that some pesticides may induce CYP2B6 via CAR-independent mechanisms. Urea 4-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 22310298-11 2012 The urea herbicide diuron and the triazine herbicide atrazine induced CYP2B6 mRNA more than 10-fold, but did not activate CAR indicating that some pesticides may induce CYP2B6 via CAR-independent mechanisms. Urea 4-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 22310298-11 2012 The urea herbicide diuron and the triazine herbicide atrazine induced CYP2B6 mRNA more than 10-fold, but did not activate CAR indicating that some pesticides may induce CYP2B6 via CAR-independent mechanisms. Diuron 19-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 22310298-11 2012 The urea herbicide diuron and the triazine herbicide atrazine induced CYP2B6 mRNA more than 10-fold, but did not activate CAR indicating that some pesticides may induce CYP2B6 via CAR-independent mechanisms. Diuron 19-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 22310298-11 2012 The urea herbicide diuron and the triazine herbicide atrazine induced CYP2B6 mRNA more than 10-fold, but did not activate CAR indicating that some pesticides may induce CYP2B6 via CAR-independent mechanisms. Triazines 34-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 22310298-11 2012 The urea herbicide diuron and the triazine herbicide atrazine induced CYP2B6 mRNA more than 10-fold, but did not activate CAR indicating that some pesticides may induce CYP2B6 via CAR-independent mechanisms. Triazines 34-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 22310298-11 2012 The urea herbicide diuron and the triazine herbicide atrazine induced CYP2B6 mRNA more than 10-fold, but did not activate CAR indicating that some pesticides may induce CYP2B6 via CAR-independent mechanisms. Atrazine 53-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 22310298-11 2012 The urea herbicide diuron and the triazine herbicide atrazine induced CYP2B6 mRNA more than 10-fold, but did not activate CAR indicating that some pesticides may induce CYP2B6 via CAR-independent mechanisms. Atrazine 53-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 22245335-4 2012 In addition, functional response of P450 systems is modulated by the presence of specific and non-specific effector molecules, metal ions, membrane incorporation, formation of homo- and hetero-oligomers, and interactions with the protein redox partners. Metals 127-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 22245335-5 2012 In this article we briefly overview the main factors contributing to the allosteric effects in cytochromes P450 with the main focus on the sources of cooperative behavior in xenobiotic metabolizing monomeric heme enzymes with their conformational flexibility and extremely broad substrate specificity. Heme 208-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-111 22245335-6 2012 The novel mechanism of functional cooperativity in P450 enzymes does not require substantial binding cooperativity, rather it implies the presence of one or more binding sites with higher affinity than the single catalytically active site in the vicinity of the heme iron. Heme 262-266 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 22245335-6 2012 The novel mechanism of functional cooperativity in P450 enzymes does not require substantial binding cooperativity, rather it implies the presence of one or more binding sites with higher affinity than the single catalytically active site in the vicinity of the heme iron. Iron 267-271 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 22281205-5 2012 While no differences in metabolite production were observed in homozygous CYP2C19*2 HLMs, homozygous CYP2B6*6 specimens produced significantly less CPF-O than wild-type specimens at 10 muM (mean 144 and 446 pmol/min/mg, respectively). cpf-o 148-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 22281205-7 2012 Additionally, CYP2B6*1 and CYP2B6*6 were over-expressed in mammalian COS-1 cells to assess for the first time the impact of the CYP2B6*6 variant on the kinetic parameters of CPF bioactivation. carbonyl sulfide 69-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 21913995-11 2012 Toxicity by CYP2B6 was abolished with the reactive oxygen species scavenger N-acetylcysteine. Reactive Oxygen Species 42-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 21913995-11 2012 Toxicity by CYP2B6 was abolished with the reactive oxygen species scavenger N-acetylcysteine. Acetylcysteine 76-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 22462748-1 2012 AIM: Hepatic enzymes, CYP2B6 and UGT2B7 play a major role in the metabolism of the widely used antiretroviral drugs efavirenz, nevirapine and zidovudine. Nevirapine 127-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 22462748-1 2012 AIM: Hepatic enzymes, CYP2B6 and UGT2B7 play a major role in the metabolism of the widely used antiretroviral drugs efavirenz, nevirapine and zidovudine. Zidovudine 142-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 21831410-7 2012 CONCLUSIONS: Apart from the CYP2C19*2, other genetic variants involved in clopidogrel metabolism and action like CYP2B6*5 and P2RY12 seem to have an important association with HTPR. Clopidogrel 74-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 22159699-0 2012 Which CYP2B6 variants have functional consequences for cyclophosphamide bioactivation? Cyclophosphamide 55-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 22687473-3 2012 One of the most successful industrial applications of P450 may be the bioconversion process for pravastatin formation using a Streptomyces carbophilus CYP105A3. Pravastatin 96-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 22190694-6 2012 An unexpected and potentially clinically significant DDI was observed between DIG and the CYP2B6 probe, bupropion, which decreased DIG AUC(0-24 h) 1.6-fold and increased Cl(renal) 1.8-fold. Digoxin 78-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 22190694-6 2012 An unexpected and potentially clinically significant DDI was observed between DIG and the CYP2B6 probe, bupropion, which decreased DIG AUC(0-24 h) 1.6-fold and increased Cl(renal) 1.8-fold. Bupropion 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 22190694-6 2012 An unexpected and potentially clinically significant DDI was observed between DIG and the CYP2B6 probe, bupropion, which decreased DIG AUC(0-24 h) 1.6-fold and increased Cl(renal) 1.8-fold. Digoxin 131-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 22022918-6 2012 This reaction was inhibited significantly by ketoconazole, a CYP3A inhibitor, and azamulin, a mechanism-based inhibitor of CYP3A, while no substantial effect was observed using selective inhibitors for eight other P450s (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP2E1). Ketoconazole 45-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 237-243 22022918-6 2012 This reaction was inhibited significantly by ketoconazole, a CYP3A inhibitor, and azamulin, a mechanism-based inhibitor of CYP3A, while no substantial effect was observed using selective inhibitors for eight other P450s (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP2E1). azamulin 82-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 237-243 22101253-4 2012 More recently, the roles of these two human ferredoxins in iron-sulfur cluster assembly were assessed, and it was concluded that FDX1 was important solely for its interaction with p450 enzymes to synthesize mitochondrial steroid precursors, whereas FDX2 was used for synthesis of iron-sulfur clusters, but not steroidogenesis. Steroids 221-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 180-184 22085411-4 2012 We synthesized caged molecules of nicotinamide adenine dinucleotide phosphate (NADP(+)) and glucose 6-phosphate (G6P), which are involved in the generation of NADPH (cofactor of P450). NADP 34-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 22085411-4 2012 We synthesized caged molecules of nicotinamide adenine dinucleotide phosphate (NADP(+)) and glucose 6-phosphate (G6P), which are involved in the generation of NADPH (cofactor of P450). NADP 79-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 22085411-4 2012 We synthesized caged molecules of nicotinamide adenine dinucleotide phosphate (NADP(+)) and glucose 6-phosphate (G6P), which are involved in the generation of NADPH (cofactor of P450). Glucose-6-Phosphate 92-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 22085411-4 2012 We synthesized caged molecules of nicotinamide adenine dinucleotide phosphate (NADP(+)) and glucose 6-phosphate (G6P), which are involved in the generation of NADPH (cofactor of P450). Glucose-6-Phosphate 113-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 22085411-4 2012 We synthesized caged molecules of nicotinamide adenine dinucleotide phosphate (NADP(+)) and glucose 6-phosphate (G6P), which are involved in the generation of NADPH (cofactor of P450). NADP 159-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 22111602-5 2012 CYP2B6 516G>T (non-wild-type/wild-type) correlated with nevirapine pharmacokinetic parameters; geometric mean ratios (95% CI): 1.75 (1.27-2.40) for area under the concentration time curve (AUC)(0-12 h), 1.58 (1.03-2.42) for C(0), and 0.53 (0.31-0.91) for clearance. Nevirapine 59-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 21976622-0 2012 Potential contribution of cytochrome P450 2B6 to hepatic 4-hydroxycyclophosphamide formation in vitro and in vivo. 4-hydroxycyclophosphamide 57-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-45 21976622-1 2012 Results from retrospective studies on the relationship between cytochrome P450 (P450) 2B6 (CYP2B6) genotype and cyclophosphamide (CY) efficacy and toxicity in adult cancer patients have been conflicting. Cyclophosphamide 112-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-89 21976622-1 2012 Results from retrospective studies on the relationship between cytochrome P450 (P450) 2B6 (CYP2B6) genotype and cyclophosphamide (CY) efficacy and toxicity in adult cancer patients have been conflicting. Cyclophosphamide 112-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 21976622-1 2012 Results from retrospective studies on the relationship between cytochrome P450 (P450) 2B6 (CYP2B6) genotype and cyclophosphamide (CY) efficacy and toxicity in adult cancer patients have been conflicting. Cyclophosphamide 91-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-89 21976622-6 2012 In human liver microsomes (HLM), 4HCY formation correlated with known phenotypic markers of CYP2B6 activity, specifically formation of (S)-2-ethyl-1,5-dimethyl-3,3-diphenyl pyrrolidine and hydroxybupropion. (s)-2-ethyl-1,5-dimethyl-3,3-diphenyl pyrrolidine 135-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 21741706-1 2012 CYP2B6, CYP2C19, ABCC4, and SOD2 have been implicated in adverse drug reactions and survival after cyclophosphamide (CPA) treatment. Cyclophosphamide 99-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 21741706-1 2012 CYP2B6, CYP2C19, ABCC4, and SOD2 have been implicated in adverse drug reactions and survival after cyclophosphamide (CPA) treatment. Cyclophosphamide 117-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 23353842-3 2012 METHODS: The chemical cross-linking by soluble carbodiimide (EDC) in combination with the liquid chromatography coupled with high resolution mass spectrometry (LC-HRMS) has been employed to characterize the contact surface regions involved in the transient interaction between two catalytic domains of P450 2B4 and cytochrome b5. Carbodiimides 47-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 302-306 23353842-3 2012 METHODS: The chemical cross-linking by soluble carbodiimide (EDC) in combination with the liquid chromatography coupled with high resolution mass spectrometry (LC-HRMS) has been employed to characterize the contact surface regions involved in the transient interaction between two catalytic domains of P450 2B4 and cytochrome b5. ethylene dichloride 61-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 302-306 22082652-14 2012 Multivariate regression analysis identified carriage of a composite genotype of ABCC10 rs2125739 and CYP2B6 516G>T (P=0.001), time post dose (P=0.01) and BMI (P=0.07) to be independently associated with nevirapine plasma concentrations. Nevirapine 206-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 21594721-3 2012 The inductive effects of tanespimycin and 17-AG on CYP1A2, CYP2B6, and CYP3A4/5 were determined in cultured primary human hepatocytes. tanespimycin 25-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 21594721-3 2012 The inductive effects of tanespimycin and 17-AG on CYP1A2, CYP2B6, and CYP3A4/5 were determined in cultured primary human hepatocytes. 17-AG 42-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 23506555-0 2012 The evaluation of CYP2B6 inhibition by artemisinin antimalarials in recombinant enzymes and human liver microsomes. artemisinin 39-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 23506555-2 2012 Some artemisinin compounds and anti-retroviral drugs have been shown to be metabolized by CYP2B6. artemisinin 5-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 23506555-4 2012 This study aimed to investigate whether artemisinin compounds inhibit CYP2B6 activity in vitro using recombinant CYP2B6 (rCYP2B6) and human liver microsomes (HLM). artemisinin 40-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 23506555-4 2012 This study aimed to investigate whether artemisinin compounds inhibit CYP2B6 activity in vitro using recombinant CYP2B6 (rCYP2B6) and human liver microsomes (HLM). artemisinin 40-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 23506555-7 2012 Artemisinin and artemether were shown to inhibit CYP2B6 in vitro through a partial mixed type of inhibition, while dihydroartemisinin did not inhibit the enzymatic activity. artemisinin 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 23506555-7 2012 Artemisinin and artemether were shown to inhibit CYP2B6 in vitro through a partial mixed type of inhibition, while dihydroartemisinin did not inhibit the enzymatic activity. Artemether 16-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 22352331-2 2012 Both CYP2B6 and CYP2C19 are pharmacologically and toxicologically relevant due to their ability to metabolize multiple drugs and environmental contaminants, including the organophosphorus (OP) pesticide chlorpyrifos. organophosphorus 171-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-11 22352331-2 2012 Both CYP2B6 and CYP2C19 are pharmacologically and toxicologically relevant due to their ability to metabolize multiple drugs and environmental contaminants, including the organophosphorus (OP) pesticide chlorpyrifos. Chlorpyrifos 203-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-11 22352331-3 2012 The aim of this study was to determine the prevalence of CYP2B6 and CYP2C19 variants in an indigenous Egyptian population (n = 120) that was shown to be occupationally exposed to chlorpyrifos. Chlorpyrifos 179-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 22573452-0 2012 Cryoradiolysis and cryospectroscopy for studies of heme-oxygen intermediates in cytochromes p450. Heme 51-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 22573452-0 2012 Cryoradiolysis and cryospectroscopy for studies of heme-oxygen intermediates in cytochromes p450. Oxygen 56-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 22573452-4 2012 Application of this method allowed for characterizing of peroxo-ferric and hydroperoxo-ferric intermediates, which are common for the oxygen activation mechanism in cytochromes P450, heme oxygenases, and nitric oxide synthases, as well as for the peroxide metabolism by peroxidases and catalases. Oxygen 134-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-181 22573452-4 2012 Application of this method allowed for characterizing of peroxo-ferric and hydroperoxo-ferric intermediates, which are common for the oxygen activation mechanism in cytochromes P450, heme oxygenases, and nitric oxide synthases, as well as for the peroxide metabolism by peroxidases and catalases. Peroxides 247-255 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-181 22111602-8 2012 The T allele for CYP2B6 516 was significantly associated with nevirapine exposure. Nevirapine 62-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 21963990-2 2011 The P450 catalytic cycle is known to uncouple and release reactive oxygen species (ROS), but the effects of ROS from P450 and other enzymes in the endoplasmic reticulum have been poorly studied from the perspective of effects on cell biology. Reactive Oxygen Species 58-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-8 21930824-0 2011 Thr302 is the site for the covalent modification of human cytochrome P450 2B6 leading to mechanism-based inactivation by tert-butylphenylacetylene. tert-butylphenylacetylene 121-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-77 21930824-1 2011 The mechanism-based inactivation of human CYP2B6 by tert-butylphenylacetylene (BPA) in the reconstituted system was investigated. tert-butylphenylacetylene 52-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 21930824-1 2011 The mechanism-based inactivation of human CYP2B6 by tert-butylphenylacetylene (BPA) in the reconstituted system was investigated. bisphenol A 79-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 21930824-2 2011 The inactivation of CYP2B6 by BPA is time-, concentration-, and NADPH-dependent and exhibits a K(I) of 2.8 muM, a k(inact) of 0.7 min(-1), and a t(1/2) of 1 min. bisphenol A 30-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 21930824-2 2011 The inactivation of CYP2B6 by BPA is time-, concentration-, and NADPH-dependent and exhibits a K(I) of 2.8 muM, a k(inact) of 0.7 min(-1), and a t(1/2) of 1 min. NADP 64-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 21930824-4 2011 Unlike CYP2B1 and CYP2B4, in addition to the formation of an apoprotein adduct and a glutathione conjugate, a small heme adduct was observed when CYP2B6 was incubated with BPA. Glutathione 85-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 146-152 21930824-4 2011 Unlike CYP2B1 and CYP2B4, in addition to the formation of an apoprotein adduct and a glutathione conjugate, a small heme adduct was observed when CYP2B6 was incubated with BPA. Heme 116-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 146-152 21930824-4 2011 Unlike CYP2B1 and CYP2B4, in addition to the formation of an apoprotein adduct and a glutathione conjugate, a small heme adduct was observed when CYP2B6 was incubated with BPA. bisphenol A 172-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 146-152 21930824-6 2011 To identify the adducted residue, BPA-inactivated CYP2B6 was digested with trypsin, and the digest was then analyzed by liquid chromatography-tandem mass spectrometry. bisphenol A 34-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 21930824-11 2011 In conclusion, Thr302 of CYP2B6 is covalently modified by a reactive metabolite of BPA, and this modification is responsible for the mechanism-based inactivation. bisphenol A 83-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 22719896-4 2012 Surprisingly, both mutants, Y181D and A287P in POR completely inhibited the CYP3A4 activity with testosterone, while the catalytic activity of CYP2B6 with bupropion was reduced to approximately ~70% of wild-type activity by Y181D and A287P mutations. Bupropion 155-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 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 2 subfamily B member 6 Homo sapiens 105-111 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Nelfinavir 164-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Ritonavir 169-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Rifampin 177-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 21963990-2 2011 The P450 catalytic cycle is known to uncouple and release reactive oxygen species (ROS), but the effects of ROS from P450 and other enzymes in the endoplasmic reticulum have been poorly studied from the perspective of effects on cell biology. Reactive Oxygen Species 83-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-8 21963990-6 2011 The antioxidant N-acetylcysteine suppressed all P450-dependent changes in protein secretion except for CD14. Acetylcysteine 16-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 21875942-0 2011 Structures of cytochrome P450 2B6 bound to 4-benzylpyridine and 4-(4-nitrobenzyl)pyridine: insight into inhibitor binding and rearrangement of active site side chains. 4-benzylpyridine 43-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-33 21209232-1 2011 Sibutramine is metabolized by the enzymes CYP2B6 and CYP2C19 into 2 active metabolites, M1 (mono-desmethyl sibutramine) and M2 (di-desmethyl sibutramine). sibutramine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 21209232-1 2011 Sibutramine is metabolized by the enzymes CYP2B6 and CYP2C19 into 2 active metabolites, M1 (mono-desmethyl sibutramine) and M2 (di-desmethyl sibutramine). sibutramine 92-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 21209232-1 2011 Sibutramine is metabolized by the enzymes CYP2B6 and CYP2C19 into 2 active metabolites, M1 (mono-desmethyl sibutramine) and M2 (di-desmethyl sibutramine). sibutramine 128-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 21209232-2 2011 Clopidogrel is a mechanism-based inhibitor of CYP2B6 and CYP2C19. Clopidogrel 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 21209232-10 2011 The CYP2B6 and CYP2C19 inhibitor clopidogrel significantly inhibited the formations of M1 from sibutramine and M2 from sibutramine by 37% and 64%, respectively. Clopidogrel 33-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 21209232-10 2011 The CYP2B6 and CYP2C19 inhibitor clopidogrel significantly inhibited the formations of M1 from sibutramine and M2 from sibutramine by 37% and 64%, respectively. sibutramine 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 21209232-10 2011 The CYP2B6 and CYP2C19 inhibitor clopidogrel significantly inhibited the formations of M1 from sibutramine and M2 from sibutramine by 37% and 64%, respectively. sibutramine 119-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 21209232-11 2011 Therefore, CYP2B6 and CYP2C19 are in vivo catalysts for the formation of the 2 active metabolites of sibutramine. sibutramine 101-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 21875942-0 2011 Structures of cytochrome P450 2B6 bound to 4-benzylpyridine and 4-(4-nitrobenzyl)pyridine: insight into inhibitor binding and rearrangement of active site side chains. 4-(4-nitrobenzyl)pyridine 64-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-33 21998091-0 2011 P450(BM3) on steroids: the Swiss Army knife P450 enzyme just gets better. Steroids 13-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-8 22054099-0 2011 Determination of cytochrome P450 enzymes involved in the metabolism of (-)-terpinen-4-ol by human liver microsomes. terpinenol-4 71-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-32 22054099-3 2011 (-)-Terpinen-4-ol was found to be oxidized to (-)-(1S,2R,4R)-1,2-epoxy-p-menthan-4-ol, major metabolic product by human liver microsomal P450 enzymes. terpinenol-4 0-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-141 22054099-3 2011 (-)-Terpinen-4-ol was found to be oxidized to (-)-(1S,2R,4R)-1,2-epoxy-p-menthan-4-ol, major metabolic product by human liver microsomal P450 enzymes. (-)-(1s,2r,4r)-1,2-epoxy-p-menthan-4-ol 46-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-141 22054099-6 2011 First, of 11 recombinant human P450 enzymes tested, CYP2A6 had the highest activity for oxidation of (-)-terpinen-4-ol. terpinenol-4 101-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 22054099-10 2011 Human recombinant CYP2A6 catalysed (-)-(1S,2R,4R)-1,2-epoxy-p-menthan-4-ol with V(max) values of 13.9 nmol/min/nmol P450 and apparent K(m) values of 91 muM. (-)-(1s,2r,4r)-1,2-epoxy-p-menthan-4-ol 35-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-120 21964418-5 2011 The substrate oxidation reactions catalyzed by CYP2A6, CYP2B6, CYP2C19 and CYP3A4 were moderately (K(i) values of 35 to 45 muM), and those by CYP1A2, CYP2D6 and CYP2E1 were weakly inhibited by PFOS (K(i) values of 190-300 muM). perfluorooctane sulfonic acid 193-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 25083217-4 2011 There is agreement amongst in vitro studies regarding the involvement of CYP1A2 and CYP2B6 in the metabolism of clopidogrel to 2-oxo-clopidogrel. Clopidogrel 112-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 25083217-4 2011 There is agreement amongst in vitro studies regarding the involvement of CYP1A2 and CYP2B6 in the metabolism of clopidogrel to 2-oxo-clopidogrel. 2-oxo-clopidogrel 127-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 21998091-2 2011 Reetz and co-workers have recently reported the development of P450(BM3) mutants that oxidise testosterone with greatly increased turnover activity, total stereoselectivity and >95 % regioselectivity. Testosterone 94-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-71 21821736-0 2011 Q172H replacement overcomes effects on the metabolism of cyclophosphamide and efavirenz caused by CYP2B6 variant with Arg262. Cyclophosphamide 57-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 21821736-0 2011 Q172H replacement overcomes effects on the metabolism of cyclophosphamide and efavirenz caused by CYP2B6 variant with Arg262. efavirenz 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 21821736-1 2011 There are a number of reports indicating that CYP2B6*6 (c.516G>T and c.785A>G) is responsible for decreased clearance of efavirenz (EFV), although increased disposition of cyclophosphamide (CPA) in individuals with this polymorphism was observed. efavirenz 138-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 21821736-1 2011 There are a number of reports indicating that CYP2B6*6 (c.516G>T and c.785A>G) is responsible for decreased clearance of efavirenz (EFV), although increased disposition of cyclophosphamide (CPA) in individuals with this polymorphism was observed. Cyclophosphamide 178-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 21821736-1 2011 There are a number of reports indicating that CYP2B6*6 (c.516G>T and c.785A>G) is responsible for decreased clearance of efavirenz (EFV), although increased disposition of cyclophosphamide (CPA) in individuals with this polymorphism was observed. Cyclophosphamide 196-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 21821736-5 2011 On the other hand, CYP2B6.6 showed a completely opposite character, suggesting that Q172H gives inverse effects on metabolic activities of CYP2B6 affected by K262R. q172h 84-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 21821736-5 2011 On the other hand, CYP2B6.6 showed a completely opposite character, suggesting that Q172H gives inverse effects on metabolic activities of CYP2B6 affected by K262R. q172h 84-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 139-145 21821736-7 2011 Furthermore, this study provides the first evidence that Q172H can reverse the direction of the effect caused by K262R in CYP2B6 on the metabolism of certain drugs. q172h 57-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 21825115-2 2011 Recently, progesterone receptor membrane component 1 (PGRMC1), which shares a key structural motif with cytochrome b(5), has been reported to bind to sterol- or steroid-synthesizing P450s, enhancing their activities. Sterols 150-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-187 21825115-2 2011 Recently, progesterone receptor membrane component 1 (PGRMC1), which shares a key structural motif with cytochrome b(5), has been reported to bind to sterol- or steroid-synthesizing P450s, enhancing their activities. Steroids 161-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-187 21825115-10 2011 In conclusion, in contrast to sterol- or steroid-synthesizing P450s, we found that PGRMC1 negatively modulates the human drug-metabolizing activities of P450 through direct interaction. Steroids 41-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-67 21825115-10 2011 In conclusion, in contrast to sterol- or steroid-synthesizing P450s, we found that PGRMC1 negatively modulates the human drug-metabolizing activities of P450 through direct interaction. Steroids 41-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. Propofol 230-238 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-67 22023129-7 2011 For all 3 species, inhibitors of CYP3A4, CYP2A6, CYP2C19, CYP2B6, and CYP2C9 diminished N-demethylation of ketamine. Nitrogen 88-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 22023129-7 2011 For all 3 species, inhibitors of CYP3A4, CYP2A6, CYP2C19, CYP2B6, and CYP2C9 diminished N-demethylation of ketamine. Ketamine 107-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 22023129-8 2011 Anti-CYP3A4, anti-CYP2C9, and anti-CYP2B6 antibodies also inhibited ketamine N-demethylation. Ketamine 68-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 22023129-13 2011 CONCLUSIONS AND CLINICAL RELEVANCE: Human-specific inhibitors of CYP2A6, CYP2C19, CYP3A4, CYP2B6, and CYP2C9 diminished ketamine N-demethylation in dogs and horses. ketamine n 120-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. Propofol 230-238 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. efavirenz 240-249 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-67 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. efavirenz 240-249 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. Bupropion 251-260 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-67 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. Bupropion 251-260 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. Mephobarbital 262-275 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-67 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. Mephobarbital 262-275 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. Propofol 285-293 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-67 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. Propofol 285-293 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. 2,6-di-sec-butyl phenol 301-324 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-67 21550074-2 2011 Prasugrel is mainly bioactivated by cytochromes P450 3A4/5 and CYP2B6. Prasugrel Hydrochloride 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 21550074-9 2011 Using recombinant microsomes, prasugrel biotransformation was mainly performed by CYP2B6, CYP2D6, CYP2C19, CYP3A4, and CYP3A5. Prasugrel Hydrochloride 30-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 21301907-1 2011 Mounting evidence thus far indicates that human cytochrome P450 2B6 (CYP2B6), an enzyme expressed at a relatively low level functionally, is primarily responsible for the metabolism of several clinically relevant drugs, including propofol, efavirenz, bupropion, mephobarbital, and the propofol analog 2,6-di-sec-butyl phenol. 2,6-di-sec-butyl phenol 301-324 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 21862689-0 2011 Mechanism-based inactivation of human cytochrome P450 2B6 by clopidogrel: involvement of both covalent modification of cysteinyl residue 475 and loss of heme. Clopidogrel 61-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-57 21550074-10 2011 With specific inhibitors of CYP3A, CYP2B6, CYP2D6, CYP2C9, and CYP2C19, active metabolite production was decreased by 38% +- 15% with 4-(4-chlorobenzyl)pyridine (CYP2B6 inhibitor) and by 45 +- 16% with ketoconazole (CYP3A inhibitor). 4-(4-Chlorobenzyl)pyridine 134-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 21550074-10 2011 With specific inhibitors of CYP3A, CYP2B6, CYP2D6, CYP2C9, and CYP2C19, active metabolite production was decreased by 38% +- 15% with 4-(4-chlorobenzyl)pyridine (CYP2B6 inhibitor) and by 45 +- 16% with ketoconazole (CYP3A inhibitor). 4-(4-Chlorobenzyl)pyridine 134-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 21550074-10 2011 With specific inhibitors of CYP3A, CYP2B6, CYP2D6, CYP2C9, and CYP2C19, active metabolite production was decreased by 38% +- 15% with 4-(4-chlorobenzyl)pyridine (CYP2B6 inhibitor) and by 45 +- 16% with ketoconazole (CYP3A inhibitor). Ketoconazole 202-214 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Prasugrel Hydrochloride 56-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Ritonavir 83-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Ritonavir 207-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 21550074-13 2011 In this in-vitro study, we found a potent inhibition of prasugrel bioactivation by ritonavir compared to the specific inhibitors of CYP3A and CYP2B6 due to the simultaneous inhibition of CYP2B6 and CYP3A by ritonavir. Ritonavir 207-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 21862689-0 2011 Mechanism-based inactivation of human cytochrome P450 2B6 by clopidogrel: involvement of both covalent modification of cysteinyl residue 475 and loss of heme. lysyl-cysteinyl-cysteinyl-arginyl-cysteinyl-lysine 119-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-57 21862689-0 2011 Mechanism-based inactivation of human cytochrome P450 2B6 by clopidogrel: involvement of both covalent modification of cysteinyl residue 475 and loss of heme. Heme 153-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-57 21862689-1 2011 We have investigated the mechanisms by which clopidogrel inactivates human cytochrome P450 2B6 (CYP2B6) in a reconstituted system. Clopidogrel 45-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-94 21862689-1 2011 We have investigated the mechanisms by which clopidogrel inactivates human cytochrome P450 2B6 (CYP2B6) in a reconstituted system. Clopidogrel 45-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 21862689-2 2011 It was found that clopidogrel and its thiolactone metabolite, 2-oxo-clopidogrel, both inactivate CYP2B6 in a time- and concentration-dependent manner. Clopidogrel 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 21862689-2 2011 It was found that clopidogrel and its thiolactone metabolite, 2-oxo-clopidogrel, both inactivate CYP2B6 in a time- and concentration-dependent manner. Thiolactone 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 21862689-2 2011 It was found that clopidogrel and its thiolactone metabolite, 2-oxo-clopidogrel, both inactivate CYP2B6 in a time- and concentration-dependent manner. 2-oxo-clopidogrel 62-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 21862689-3 2011 On the basis of k(inact)/K(I) ratios, clopidogrel is approximately 5 times more efficient than 2-oxo-clopidogrel in inactivating CYP2B6. Clopidogrel 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 21862689-3 2011 On the basis of k(inact)/K(I) ratios, clopidogrel is approximately 5 times more efficient than 2-oxo-clopidogrel in inactivating CYP2B6. 2-oxo-clopidogrel 95-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 21862689-4 2011 Analysis of the molecular mass of the CYP2B6 wild-type (WT) protein that had been inactivated by either clopidogrel or 2-oxo-clopidogrel showed an increase in the mass of the protein by ~350 Da. Clopidogrel 104-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 21862689-4 2011 Analysis of the molecular mass of the CYP2B6 wild-type (WT) protein that had been inactivated by either clopidogrel or 2-oxo-clopidogrel showed an increase in the mass of the protein by ~350 Da. 2-oxo-clopidogrel 119-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 21862689-5 2011 This increase in the protein mass corresponds to the addition of the active metabolite of clopidogrel to CYP2B6. Clopidogrel 90-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 21862689-6 2011 It is noteworthy that this adduct can be cleaved from the protein matrix by incubation with dithiothreitol, confirming that the active metabolite is linked to a cysteinyl residue of CYP2B6 via a disulfide bond. Dithiothreitol 92-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-188 21862689-6 2011 It is noteworthy that this adduct can be cleaved from the protein matrix by incubation with dithiothreitol, confirming that the active metabolite is linked to a cysteinyl residue of CYP2B6 via a disulfide bond. lysyl-cysteinyl-cysteinyl-arginyl-cysteinyl-lysine 161-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-188 21862689-6 2011 It is noteworthy that this adduct can be cleaved from the protein matrix by incubation with dithiothreitol, confirming that the active metabolite is linked to a cysteinyl residue of CYP2B6 via a disulfide bond. Disulfides 195-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-188 21862689-7 2011 Peptide mapping of tryptic digests of the inactivated CYP2B6 using electrospray ionization liquid chromatography-tandem mass spectrometry identified Cys475 as the site of covalent modification by the active metabolite. Peptides 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 21862689-10 2011 Furthermore, inactivation of both CYP2B6 WT and C475S by clopidogrel, but not by 2-oxo-clopidogrel, led to the loss of the heme, which accounts for most of the loss of the catalytic activity. Clopidogrel 57-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 21862689-10 2011 Furthermore, inactivation of both CYP2B6 WT and C475S by clopidogrel, but not by 2-oxo-clopidogrel, led to the loss of the heme, which accounts for most of the loss of the catalytic activity. Heme 123-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 21862689-11 2011 Collectively, these results suggest that clopidogrel inactivates CYP2B6 primarily through destruction of the heme, whereas 2-oxo-clopidogrel inactivates CYP2B6 through covalent modification of Cys475. Clopidogrel 41-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 21862689-11 2011 Collectively, these results suggest that clopidogrel inactivates CYP2B6 primarily through destruction of the heme, whereas 2-oxo-clopidogrel inactivates CYP2B6 through covalent modification of Cys475. Heme 109-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 21862689-11 2011 Collectively, these results suggest that clopidogrel inactivates CYP2B6 primarily through destruction of the heme, whereas 2-oxo-clopidogrel inactivates CYP2B6 through covalent modification of Cys475. 2-oxo-clopidogrel 123-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 21846671-8 2011 The Wilcoxon matched pairs test indicated that the change in 8-hydroxyefavirenz concentration and efavirenz MR over time was significant in females and in CYP2B6*1 and UGT2B7*1 carriers. 8-hydroxyefavirenz 61-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-161 21902500-3 2011 RESULTS: Pair-wise comparisons revealed that carriers of the variants ABCB1 3435C>T or CYP2B6 516G>T alleles were more likely to require a higher methadone dose than noncarriers (both p < 0.0001). Methadone 152-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 21902500-5 2011 Proportional odds regression with adjustment of cofactors demonstrated that ABCB1, CYP2B6, OPRM1, ANKK1 and DRD2 genetic variants were jointly correlated with optimal methadone dose (adjusted r(2) = 53%). Methadone 167-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 21839818-3 2011 It has been reported that duloxetine causes time-dependent inhibition (TDI) of CYP1A2, CYP2B6, CYP2C19 and CYP3A4/5; but the nature of these TDI (whether reversible or irreversible) is not known. Duloxetine Hydrochloride 26-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 21860339-11 2011 CONCLUSION: By integration of high-throughput genotyping data into a pharmacometric analysis of nevirapine, the impact of the CYP2B6 516G>T polymorphism on nevirapine"s exposure was confirmed and quantified. Nevirapine 96-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 21746968-0 2011 Functional characterization of CYP2B6 allelic variants in demethylation of antimalarial artemether. Artemether 88-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 21746968-4 2011 Of 14 recombinant P450s examined in this study, CYP2B6 and CYP3A4 were primarily responsible for production of the desmethyl metabolite dihydroartemisinin. artenimol 136-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-54 21737533-4 2011 P450 functions can be categorized in several groups: 1) Some P450s have critical roles in the metabolism of endogenous substrates (e.g., sterols and fat-soluble vitamins). Sterols 137-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 21726172-5 2011 However, toremifene is a competitive inhibitor of CYP3A4, non-competitive inhibitor of CYP2A6, 2C8, 2C9, 2C19 and 2E1 and mixed-type inhibitor of CYP2B6. Toremifene 9-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 146-152 21726172-8 2011 Although clinical doses of toremifene produce steady state exposures to toremifene and NDMT that may be sufficient to cause pharmacokinetic drug-drug interactions with other drugs metabolised by CYP2B6, CYP2C8, CYP3A4, CYP2C9 and CYP2C19, these data indicate that toremifene is unlikely to play a role in clinical drug-drug interactions with substrate drugs of CYP1A2 and CYP2D6. Toremifene 27-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-201 21726172-8 2011 Although clinical doses of toremifene produce steady state exposures to toremifene and NDMT that may be sufficient to cause pharmacokinetic drug-drug interactions with other drugs metabolised by CYP2B6, CYP2C8, CYP3A4, CYP2C9 and CYP2C19, these data indicate that toremifene is unlikely to play a role in clinical drug-drug interactions with substrate drugs of CYP1A2 and CYP2D6. N-desmethyltoremifene 87-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-201 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 2 subfamily B member 6 Homo sapiens 189-195 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 2 subfamily B member 6 Homo sapiens 189-195 21871151-7 2011 Individual susceptibility to methadone may be determined by screening for CYP2B6*6. Methadone 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 21659470-0 2011 Polymorphic variants of cytochrome P450 2B6 (CYP2B6.4-CYP2B6.9) exhibit altered rates of metabolism for bupropion and efavirenz: a charge-reversal mutation in the K139E variant (CYP2B6.8) impairs formation of a functional cytochrome p450-reductase complex. Bupropion 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 21659470-0 2011 Polymorphic variants of cytochrome P450 2B6 (CYP2B6.4-CYP2B6.9) exhibit altered rates of metabolism for bupropion and efavirenz: a charge-reversal mutation in the K139E variant (CYP2B6.8) impairs formation of a functional cytochrome p450-reductase complex. Bupropion 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 21659470-0 2011 Polymorphic variants of cytochrome P450 2B6 (CYP2B6.4-CYP2B6.9) exhibit altered rates of metabolism for bupropion and efavirenz: a charge-reversal mutation in the K139E variant (CYP2B6.8) impairs formation of a functional cytochrome p450-reductase complex. Bupropion 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. Bupropion 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. Bupropion 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. Bupropion 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. Bupropion 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. efavirenz 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. efavirenz 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. efavirenz 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. efavirenz 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. 7-ethoxy-4-trifluoromethylcoumarin 55-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. 7-ethoxy-4-trifluoromethylcoumarin 55-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. 7-ethoxy-4-trifluoromethylcoumarin 55-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 21659470-1 2011 In this study, metabolism of bupropion, efavirenz, and 7-ethoxy-4-trifluoromethylcoumarin (7-EFC) by CYP2B6 wild type (CYP2B6.1) and six polymorphic variants (CYP2B6.4 to CYP2B6.9) was investigated in a reconstituted system to gain a better understanding of the effects of the mutations on the catalytic properties of these naturally occurring variants. 7-ethoxy-4-trifluoromethylcoumarin 55-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 21659470-3 2011 The steady-state turnover rates for the hydroxylation of bupropion and efavirenz and the O-deethylation of 7-EFC showed that these mutations significantly alter the catalytic activities of CYP2B6. Bupropion 57-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-195 21659470-3 2011 The steady-state turnover rates for the hydroxylation of bupropion and efavirenz and the O-deethylation of 7-EFC showed that these mutations significantly alter the catalytic activities of CYP2B6. efavirenz 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-195 21659470-4 2011 It was found that CYP2B6.6 exhibits 4- and 27-fold increases in the K(m) values for the hydroxylation of bupropion and efavirenz, respectively, and CYP2B6.8 completely loses its ability to metabolize any of the substrates under normal turnover conditions. Bupropion 105-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 21659470-4 2011 It was found that CYP2B6.6 exhibits 4- and 27-fold increases in the K(m) values for the hydroxylation of bupropion and efavirenz, respectively, and CYP2B6.8 completely loses its ability to metabolize any of the substrates under normal turnover conditions. Bupropion 105-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-154 21659470-4 2011 It was found that CYP2B6.6 exhibits 4- and 27-fold increases in the K(m) values for the hydroxylation of bupropion and efavirenz, respectively, and CYP2B6.8 completely loses its ability to metabolize any of the substrates under normal turnover conditions. efavirenz 119-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 21659470-4 2011 It was found that CYP2B6.6 exhibits 4- and 27-fold increases in the K(m) values for the hydroxylation of bupropion and efavirenz, respectively, and CYP2B6.8 completely loses its ability to metabolize any of the substrates under normal turnover conditions. efavirenz 119-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-154 21659470-5 2011 However, compared with CYP2B6.1, CYP2B6.8 retains 77% of its 7-EFC O-deethylase activity in the presence of tert-butyl hydroperoxide as an alternative oxidant, indicating that the heme and the active site are catalytically competent. tert-Butylhydroperoxide 108-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 21659470-5 2011 However, compared with CYP2B6.1, CYP2B6.8 retains 77% of its 7-EFC O-deethylase activity in the presence of tert-butyl hydroperoxide as an alternative oxidant, indicating that the heme and the active site are catalytically competent. Heme 180-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 21659470-6 2011 Presteady-state measurements of the rate of electron transfer from NADPH-dependent cytochrome P450 reductase (CPR) to CYP2B6.8 using stopped-flow spectrophotometry revealed that CYP2B6.8 is incapable of accepting electrons from CPR. NADP 67-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 21659470-6 2011 Presteady-state measurements of the rate of electron transfer from NADPH-dependent cytochrome P450 reductase (CPR) to CYP2B6.8 using stopped-flow spectrophotometry revealed that CYP2B6.8 is incapable of accepting electrons from CPR. NADP 67-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-184 21902501-2 2011 The isozyme CYP3A4 of the CYP system is one of the metabolic enzymes, as well as CYP2B6, responsible for the metabolism of methadone. Methadone 123-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 21906462-4 2011 TAM is known to be metabolised to its active metabolites N-desmethyl TAM and 4-hydroxytamoxifen by a number of CYP450 enzymes, including CYP2D6, CYP3A4, CYP2C9, CYP2C19 and CYP2B6. Tamoxifen 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 21906462-4 2011 TAM is known to be metabolised to its active metabolites N-desmethyl TAM and 4-hydroxytamoxifen by a number of CYP450 enzymes, including CYP2D6, CYP3A4, CYP2C9, CYP2C19 and CYP2B6. N-desmethyltamoxifen 57-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 21906462-4 2011 TAM is known to be metabolised to its active metabolites N-desmethyl TAM and 4-hydroxytamoxifen by a number of CYP450 enzymes, including CYP2D6, CYP3A4, CYP2C9, CYP2C19 and CYP2B6. hydroxytamoxifen 77-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 21659470-0 2011 Polymorphic variants of cytochrome P450 2B6 (CYP2B6.4-CYP2B6.9) exhibit altered rates of metabolism for bupropion and efavirenz: a charge-reversal mutation in the K139E variant (CYP2B6.8) impairs formation of a functional cytochrome p450-reductase complex. Bupropion 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-43 21555506-12 2011 Summing up, we showed that, whereas liver P450 isoenzymes were involved in the metabolism of C-1311 to a limited extent, FMO plays a significant role in the microsomal transformations of this compound, which is also a specific substrate of UGT1A1. C 1311 93-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 21716267-1 2011 This study examined drug-drug interactions of oral S-ketamine with the cytochrome P450 (CYP) 2B6 inhibitor ticlopidine and the CYP3A inhibitor itraconazole. Ketamine 51-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-96 21716267-1 2011 This study examined drug-drug interactions of oral S-ketamine with the cytochrome P450 (CYP) 2B6 inhibitor ticlopidine and the CYP3A inhibitor itraconazole. Ticlopidine 107-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-96 21210253-9 2011 Finally we showed that lentivirus-mediated expression of xenoreceptor CAR in ES-Hep induced the expression of several detoxification genes including CYP2B6, CYP2C9, CYP3A4, UDP-glycosyltransferase 1A1, solute carriers 21A6, as well as biotransformation of midazolam, a CYP3A4-specific substrate. Midazolam 256-265 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 21570381-5 2011 The selective inhibitors of CYP2B6 (ticlopidine and phencyclidine) and monoclonal antibodies raised against human CYP2B6 isozyme caused 80% inhibition of OH-BUP formation by baboon hepatic microsomes. Ticlopidine 36-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 21570381-5 2011 The selective inhibitors of CYP2B6 (ticlopidine and phencyclidine) and monoclonal antibodies raised against human CYP2B6 isozyme caused 80% inhibition of OH-BUP formation by baboon hepatic microsomes. Phencyclidine 52-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 21570381-5 2011 The selective inhibitors of CYP2B6 (ticlopidine and phencyclidine) and monoclonal antibodies raised against human CYP2B6 isozyme caused 80% inhibition of OH-BUP formation by baboon hepatic microsomes. oh-bup 154-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 21570381-5 2011 The selective inhibitors of CYP2B6 (ticlopidine and phencyclidine) and monoclonal antibodies raised against human CYP2B6 isozyme caused 80% inhibition of OH-BUP formation by baboon hepatic microsomes. oh-bup 154-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 21694616-0 2011 CYP2B6 polymorphisms influence the plasma concentration and clearance of the methadone S-enantiomer. Methadone 77-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 21679153-5 2011 Among recombinant P450 and FMO enzymes; CYP2D6 had a high activity in terms of tyramine elimination. Tyramine 79-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-30 21694616-10 2011 We conclude that genetic polymorphisms in the CYP2B6 gene may therefore be indicators of the clearance, plasma concentration and C/D ratio of S-methadone. Methadone 142-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 21487929-4 2011 RESULTS: CYP2B6 reporter activity was significantly enhanced by CPA and IFO in HepG2 cells co-transfected with CYP2B6 reporter plasmid and a chemical-responsive human CAR variant (CAR1 + A) construct. Cyclophosphamide 64-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 21487929-4 2011 RESULTS: CYP2B6 reporter activity was significantly enhanced by CPA and IFO in HepG2 cells co-transfected with CYP2B6 reporter plasmid and a chemical-responsive human CAR variant (CAR1 + A) construct. Cyclophosphamide 64-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 21487929-5 2011 Real-time RT-PCR and Western blotting analysis in HPHs showed that both CPA and IFO induced the expressions of CYP2B6 and CYP3A4. Cyclophosphamide 72-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 21487929-7 2011 Further studies in HPHs demonstrated that selective inhibition of PXR by sulforaphane preferentially repressed IFO- over CPA-mediated induction of CYP2B6. sulforaphane 73-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 21487929-7 2011 Further studies in HPHs demonstrated that selective inhibition of PXR by sulforaphane preferentially repressed IFO- over CPA-mediated induction of CYP2B6. Cyclophosphamide 121-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 21514354-6 2011 PBPK/PD model simulations estimated that a 4-fold increase or decrease in relative CYP2B6 and CYP2C19 content would produce a 9-22% inhibition in blood AChE activity following exposure of an adult to chlorpyrifos (1000 mug/kg). Chlorpyrifos 200-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 21514354-9 2011 Changes in hepatic CYP2B6 and CYP2C19 content had more of an influence on cholinesterase inhibition for exposures to chlorpyrifos than parathion, which agrees with previously reported literature that these CYPs are more reaction biased for desulfurization (activation) and dearylation (detoxification) of chlorpyrifos compared to parathion. Chlorpyrifos 117-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 21514354-2 2011 Phosphororthioate OPs like chlorpyrifos and parathion are directly activated and detoxified by various cytochrome P450s (CYPs), with the primary CYPs involved being CYP2B6 and CYP2C19. phosphororthioate 0-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-171 21514354-9 2011 Changes in hepatic CYP2B6 and CYP2C19 content had more of an influence on cholinesterase inhibition for exposures to chlorpyrifos than parathion, which agrees with previously reported literature that these CYPs are more reaction biased for desulfurization (activation) and dearylation (detoxification) of chlorpyrifos compared to parathion. Parathion 135-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 21514354-9 2011 Changes in hepatic CYP2B6 and CYP2C19 content had more of an influence on cholinesterase inhibition for exposures to chlorpyrifos than parathion, which agrees with previously reported literature that these CYPs are more reaction biased for desulfurization (activation) and dearylation (detoxification) of chlorpyrifos compared to parathion. Chlorpyrifos 305-317 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 21514354-2 2011 Phosphororthioate OPs like chlorpyrifos and parathion are directly activated and detoxified by various cytochrome P450s (CYPs), with the primary CYPs involved being CYP2B6 and CYP2C19. OPS 18-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-171 21514354-2 2011 Phosphororthioate OPs like chlorpyrifos and parathion are directly activated and detoxified by various cytochrome P450s (CYPs), with the primary CYPs involved being CYP2B6 and CYP2C19. Chlorpyrifos 27-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-171 21514354-2 2011 Phosphororthioate OPs like chlorpyrifos and parathion are directly activated and detoxified by various cytochrome P450s (CYPs), with the primary CYPs involved being CYP2B6 and CYP2C19. Parathion 44-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-171 21514354-9 2011 Changes in hepatic CYP2B6 and CYP2C19 content had more of an influence on cholinesterase inhibition for exposures to chlorpyrifos than parathion, which agrees with previously reported literature that these CYPs are more reaction biased for desulfurization (activation) and dearylation (detoxification) of chlorpyrifos compared to parathion. Parathion 330-339 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 21393201-3 2011 OBJECTIVES: To study the influence of age, drug dose and formulation type, nutritional status and CYP2B6 516G>T polymorphism on blood concentrations of nevirapine in children treated with generic antiretroviral drugs. Nevirapine 155-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 21436404-6 2011 Ketoconazole treatment decreased the CYP3A4 activity by 69%, and rifampicin induced the CYP3A4- and CYP2B6-dependent activity 6-fold, which predicts well the magnitude of changes observed in vivo. Rifampin 65-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 21698750-3 2011 The known activities of a small number of the 13,000 members of the P450 superfamily fall into two general classes: promiscuous enzymes that are not considered as moonlighting and forms that participate in biosynthesis of endogenous compounds, such as steroids, vitamins and play different roles in different tissues, sometimes being moonlighting enzymes. Steroids 252-260 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 21287405-8 2011 may cause food-drug interactions via cytochrome P450 inhibition by sesquiterpene 7 and curcuminoids 11 and 12. Sesquiterpenes 67-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 21287405-8 2011 may cause food-drug interactions via cytochrome P450 inhibition by sesquiterpene 7 and curcuminoids 11 and 12. curcuminoids 87-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 21508826-10 2011 CONCLUSIONS: : A novel observation is the large effect of rifampicin on S-norketamine concentrations and indicates that rifampicin induces the elimination of S-ketamine"s metabolite, S-norketamine, probably via induction of the CYP3A4 and/or CYP2B6 enzymes. Rifampin 120-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-248 21508826-10 2011 CONCLUSIONS: : A novel observation is the large effect of rifampicin on S-norketamine concentrations and indicates that rifampicin induces the elimination of S-ketamine"s metabolite, S-norketamine, probably via induction of the CYP3A4 and/or CYP2B6 enzymes. Ketamine 158-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-248 21508826-10 2011 CONCLUSIONS: : A novel observation is the large effect of rifampicin on S-norketamine concentrations and indicates that rifampicin induces the elimination of S-ketamine"s metabolite, S-norketamine, probably via induction of the CYP3A4 and/or CYP2B6 enzymes. norketamine 183-196 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-248 21441248-1 2011 BACKGROUND: Nevirapine is metabolized by CYP2B6 and polymorphisms within the CYP2B6 gene partly explain inter-patient variability in pharmacokinetics. Nevirapine 12-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 21469227-1 2011 Cytochromes P450 catalyze a range of different oxygen-transfer processes including aliphatic and aromatic hydroxylation, epoxidation, and sulfoxidation reactions. Oxygen 47-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-16 21441248-1 2011 BACKGROUND: Nevirapine is metabolized by CYP2B6 and polymorphisms within the CYP2B6 gene partly explain inter-patient variability in pharmacokinetics. Nevirapine 12-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 21441248-2 2011 The aim of this study was to model the complex relationship between nevirapine exposure, weight and genetics (based on combined analysis of CYP2B6 516G > T and 983T > C single nucleotide polymorphisms). Nevirapine 68-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 21543025-6 2011 This investigation was done to determine the impact of CYP2B6*5 and CYP2C19*2 on the renal response in cyclophosphamide-treated lupus nephritis (LN) patients. Cyclophosphamide 103-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 21462923-4 2011 Specifically, in order to study P450--CPR interactions, a single reactive cysteine was introduced in to a genetically engineered variant of CYP2B4 (C79SC152S) at each of seven strategically selected surface-exposed positions. Cysteine 74-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 21469680-5 2011 The most relevant human drug metabolizing cytochromes P450, isoforms 3A4, 2D6, and 2C9, have been covalently bound to a gold electrode via a 10-carboxydecanethiol and 8-hydroxyoctanethiol (1:1) self-assembled monolayer at the bottom of an eight-well microtiter plate. 10-carboxydecanethiol 141-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-77 21469680-5 2011 The most relevant human drug metabolizing cytochromes P450, isoforms 3A4, 2D6, and 2C9, have been covalently bound to a gold electrode via a 10-carboxydecanethiol and 8-hydroxyoctanethiol (1:1) self-assembled monolayer at the bottom of an eight-well microtiter plate. 8-hydroxyoctanethiol 167-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-77 21395287-2 2011 When incubated in the presence of P450 and NADPH, the anticipated phenyl acetic acid metabolite was formed. phenylacetic acid 66-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-48 21289075-3 2011 Human CYP2B6 and CYP2D6, which are expressed heterogeneously throughout the brain, exhibit clinically significant polymorphisms and are regulated by external factors, such as alcohol and smoking. Alcohols 175-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 21185724-3 2011 Tamoxifen is a pro-drug that is metabolised to its active metabolites by the cytochrome P450 (CYP) enzymes including CYP2D6, CYP3A, CYP2B6, and CYP2C19. Tamoxifen 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-138 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Rifampin 116-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 236-242 21111054-4 2011 METHODS: To evaluate the xenobiotic-mediated induction of hepatocellular gene expression, the prototypical inducers rifampicin (10 muM) and phenobarbital (1 mM) were used for CYP3A4, CITCO (1 muM) and artemisinin (50 muM) were used for CYP2B6, and 3-methylcholanthrene (1 muM) and omeprazole (50 muM) were utilized for induction of CYP1A2. Phenobarbital 140-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 236-242 21442647-0 2011 The importance of correct assignment of CYP2B6 genetic variants with respect to cyclophosphamide metabolizer status. Cyclophosphamide 80-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 21395646-0 2011 In silico prediction of efavirenz and rifampicin drug-drug interaction considering weight and CYP2B6 phenotype. efavirenz 24-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 21395646-0 2011 In silico prediction of efavirenz and rifampicin drug-drug interaction considering weight and CYP2B6 phenotype. Rifampin 38-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 21395646-4 2011 The model was used to simulate the effects of rifampicin on efavirenz pharmacokinetics in 400 virtual patients, taking into account bodyweight and CYP2B6 phenotype. Rifampin 46-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 21395646-8 2011 Increasing the efavirenz dose during concomitant rifampicin was predicted to be most successful in patients over 50 kg regardless of CYP2B6 status. efavirenz 15-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 21434664-1 2011 An assaying method of cytochrome P450 (P450 or CYP) monooxygenase activities for toxicological evaluation of drugs and environmental pollutants was developed by immobilizing P450 on an oxygen sensoring layer. Oxygen 56-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-50 21185724-11 2011 This review article will summarize the available published breast cancer data on the interaction between the relevant SNPs for CYP2D6, CYP3A, CYP2B6, and CYP2C19 and the efficacy of tamoxifen, their role in individualisation of hormonal therapy and the role of the commercially available genotyping kits. Tamoxifen 182-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 20851096-9 2011 Catalytic turnover of P450(cin) with either of these alternative substrates (camphane or cinane) revealed that in the absence of the ethereal oxygen there was still a significant amount of coupling of the NADPH-reducing equivalents to the formation of oxidised product. camphane 77-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 21308129-4 2011 Although P450(BSbeta) exclusively catalyzes peroxygenation of long-alkyl-chain fatty acids, oxidation of non-natural substrates such as styrene, ethylbenzene, and 1-methoxynaphthalen are catalyzed by P450(BSbeta) in the presence of decoy molecules having a carboxyl group. long-alkyl-chain fatty acids 62-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-21 21308129-4 2011 Although P450(BSbeta) exclusively catalyzes peroxygenation of long-alkyl-chain fatty acids, oxidation of non-natural substrates such as styrene, ethylbenzene, and 1-methoxynaphthalen are catalyzed by P450(BSbeta) in the presence of decoy molecules having a carboxyl group. long-alkyl-chain fatty acids 62-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 200-212 21308129-4 2011 Although P450(BSbeta) exclusively catalyzes peroxygenation of long-alkyl-chain fatty acids, oxidation of non-natural substrates such as styrene, ethylbenzene, and 1-methoxynaphthalen are catalyzed by P450(BSbeta) in the presence of decoy molecules having a carboxyl group. Styrene 136-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 200-212 21308129-4 2011 Although P450(BSbeta) exclusively catalyzes peroxygenation of long-alkyl-chain fatty acids, oxidation of non-natural substrates such as styrene, ethylbenzene, and 1-methoxynaphthalen are catalyzed by P450(BSbeta) in the presence of decoy molecules having a carboxyl group. ethylbenzene 145-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 200-212 21308129-4 2011 Although P450(BSbeta) exclusively catalyzes peroxygenation of long-alkyl-chain fatty acids, oxidation of non-natural substrates such as styrene, ethylbenzene, and 1-methoxynaphthalen are catalyzed by P450(BSbeta) in the presence of decoy molecules having a carboxyl group. 1-methoxynaphthalen 163-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-21 21308129-4 2011 Although P450(BSbeta) exclusively catalyzes peroxygenation of long-alkyl-chain fatty acids, oxidation of non-natural substrates such as styrene, ethylbenzene, and 1-methoxynaphthalen are catalyzed by P450(BSbeta) in the presence of decoy molecules having a carboxyl group. 1-methoxynaphthalen 163-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 200-212 21308129-7 2011 The crystal structure analysis of the decoy molecule bound-form of P450(BSbeta) shows that P450(BSbeta) accepts the decoy molecule, whose carboxylate is located at the same position to that of long-alkyl-chain fatty acid. carboxylate 138-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-79 21308129-7 2011 The crystal structure analysis of the decoy molecule bound-form of P450(BSbeta) shows that P450(BSbeta) accepts the decoy molecule, whose carboxylate is located at the same position to that of long-alkyl-chain fatty acid. carboxylate 138-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-103 21308129-7 2011 The crystal structure analysis of the decoy molecule bound-form of P450(BSbeta) shows that P450(BSbeta) accepts the decoy molecule, whose carboxylate is located at the same position to that of long-alkyl-chain fatty acid. long-alkyl-chain fatty acid 193-220 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-79 21308129-7 2011 The crystal structure analysis of the decoy molecule bound-form of P450(BSbeta) shows that P450(BSbeta) accepts the decoy molecule, whose carboxylate is located at the same position to that of long-alkyl-chain fatty acid. long-alkyl-chain fatty acid 193-220 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-103 21227907-8 2011 Ex vivo studies showed DEHP and di-isononyl phthalate potently induced CYP2B6 and CYP3A4 expression in human hepatocytes. Diethylhexyl Phthalate 23-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 21227907-8 2011 Ex vivo studies showed DEHP and di-isononyl phthalate potently induced CYP2B6 and CYP3A4 expression in human hepatocytes. diisononyl phthalate 32-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 20851096-0 2011 Oxygen activation by P450(cin): Protein and substrate mutagenesis. Oxygen 0-6 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 20851096-1 2011 A conserved threonine found in the majority of cytochromes P450 (P450s) has been implicated in the activation of dioxygen during the catalytic cycle. Threonine 12-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-63 20851096-1 2011 A conserved threonine found in the majority of cytochromes P450 (P450s) has been implicated in the activation of dioxygen during the catalytic cycle. Oxygen 113-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-63 20851096-9 2011 Catalytic turnover of P450(cin) with either of these alternative substrates (camphane or cinane) revealed that in the absence of the ethereal oxygen there was still a significant amount of coupling of the NADPH-reducing equivalents to the formation of oxidised product. cinane 89-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 20851096-2 2011 P450(cin) (CYP176A) has been found to be an exception to this paradigm, where the conserved threonine has been replaced with an asparagine. Threonine 92-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 20851096-2 2011 P450(cin) (CYP176A) has been found to be an exception to this paradigm, where the conserved threonine has been replaced with an asparagine. Asparagine 128-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 20851096-3 2011 Prior studies with a P450(cin) N242A mutant established that the Asn-242 was not a functional replacement for the conserved threonine but was essential for the regio- and stereocontrol of the oxidation of cineole. Asparagine 65-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 20851096-9 2011 Catalytic turnover of P450(cin) with either of these alternative substrates (camphane or cinane) revealed that in the absence of the ethereal oxygen there was still a significant amount of coupling of the NADPH-reducing equivalents to the formation of oxidised product. Oxygen 142-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 20851096-9 2011 Catalytic turnover of P450(cin) with either of these alternative substrates (camphane or cinane) revealed that in the absence of the ethereal oxygen there was still a significant amount of coupling of the NADPH-reducing equivalents to the formation of oxidised product. NADP 205-210 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 20851096-3 2011 Prior studies with a P450(cin) N242A mutant established that the Asn-242 was not a functional replacement for the conserved threonine but was essential for the regio- and stereocontrol of the oxidation of cineole. Eucalyptol 205-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 20851096-11 2011 Thus, it still remains unclear how dioxygen activation in the catalytic turnover of cineole by P450(cin) is controlled. Oxygen 35-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 20851096-4 2011 To explore further how P450(cin) controls the activation of the dioxygen in the absence of the conserved threonine, two concurrent lines of investigation were followed. Oxygen 64-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 20851096-11 2011 Thus, it still remains unclear how dioxygen activation in the catalytic turnover of cineole by P450(cin) is controlled. Eucalyptol 84-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 20920462-3 2011 Plant P450s catalyze a wide variety of monooxygenation/hydroxylation reactions in secondary metabolism, and some of them are involved in unusual reactions such as methylenedioxy-bridge formation, phenol coupling reactions, oxidative rearrangement of carbon skeletons, and oxidative C-C bond cleavage. Phenol 196-202 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-11 20920462-3 2011 Plant P450s catalyze a wide variety of monooxygenation/hydroxylation reactions in secondary metabolism, and some of them are involved in unusual reactions such as methylenedioxy-bridge formation, phenol coupling reactions, oxidative rearrangement of carbon skeletons, and oxidative C-C bond cleavage. Carbon 250-256 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-11 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 2 subfamily B member 6 Homo sapiens 185-191 21156812-0 2011 Comparison of in vitro metabolism of ticlopidine by human cytochrome P450 2B6 and rabbit cytochrome P450 2B4. Ticlopidine 37-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-77 21336516-7 2011 The most selective inhibitors available are furafylline (in co-incubation and pre-incubation conditions) for CYP1A2, 2-phenyl-2-(1-piperidinyl)propane (PPP) for CYP2B6, montelukast for CYP2C8, sulfaphenazole for CYP2C9, (-)-N-3-benzyl-phenobarbital for CYP2C19 and quinidine for CYP2D6. furafylline 44-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 21081644-8 2011 These data show that in contrast to sterol synthesizing P450, PGRMC1 is not required for the activities of several drug-metabolizing P450s, and its overexpression inhibits those P450 activities. Sterols 36-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 21280577-3 2011 For the structural and biochemical basis of this unparalleled sequence, four crystal structures of AurH variants in different conformational states and in complex with the P450 inhibitor ancymidol were solved, which represent the first structures of the CYP151A group. ancymidol 187-196 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 21165551-2 2011 Here, we have utilised the tumour-infiltrating properties of macrophages as vehicles to deliver a gene encoding a prodrug-activating enzyme such as human cytochrome P450 2B6 (CYP2B6) inside tumours followed by killing the tumour cells with the prodrug cyclophosphamide (CPA). Cyclophosphamide 252-268 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-173 21150552-0 2011 Paradoxically elevated efavirenz concentrations in HIV/tuberculosis-coinfected patients with CYP2B6 516TT genotype on rifampin-containing antituberculous therapy. Rifampin 118-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 21150552-3 2011 Two-way analysis of variance revealed a significant interaction between CYP2B6 516G T polymorphism and rifampin-containing therapy, suggesting that efavirenz dose adjustment may need to be individualized on the basis of the patient"s genotype. Rifampin 103-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 21148250-5 2011 Dibenzylfluorescein (DBF) is widely used as a profluorescent probe substrate for P450 activity and inhibition assays, but its use has been considered to be limited to traditional endpoint assays. dibenzylfluorescein 0-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 21148250-5 2011 Dibenzylfluorescein (DBF) is widely used as a profluorescent probe substrate for P450 activity and inhibition assays, but its use has been considered to be limited to traditional endpoint assays. dibenzylfluorescein 21-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 21336516-7 2011 The most selective inhibitors available are furafylline (in co-incubation and pre-incubation conditions) for CYP1A2, 2-phenyl-2-(1-piperidinyl)propane (PPP) for CYP2B6, montelukast for CYP2C8, sulfaphenazole for CYP2C9, (-)-N-3-benzyl-phenobarbital for CYP2C19 and quinidine for CYP2D6. 2-phenyl-2-(1-piperidinyl)propane 152-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 21336516-7 2011 The most selective inhibitors available are furafylline (in co-incubation and pre-incubation conditions) for CYP1A2, 2-phenyl-2-(1-piperidinyl)propane (PPP) for CYP2B6, montelukast for CYP2C8, sulfaphenazole for CYP2C9, (-)-N-3-benzyl-phenobarbital for CYP2C19 and quinidine for CYP2D6. montelukast 169-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 21336516-7 2011 The most selective inhibitors available are furafylline (in co-incubation and pre-incubation conditions) for CYP1A2, 2-phenyl-2-(1-piperidinyl)propane (PPP) for CYP2B6, montelukast for CYP2C8, sulfaphenazole for CYP2C9, (-)-N-3-benzyl-phenobarbital for CYP2C19 and quinidine for CYP2D6. Sulfaphenazole 193-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 21336516-7 2011 The most selective inhibitors available are furafylline (in co-incubation and pre-incubation conditions) for CYP1A2, 2-phenyl-2-(1-piperidinyl)propane (PPP) for CYP2B6, montelukast for CYP2C8, sulfaphenazole for CYP2C9, (-)-N-3-benzyl-phenobarbital for CYP2C19 and quinidine for CYP2D6. N-3-benzylphenobarbital 220-248 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 21336516-7 2011 The most selective inhibitors available are furafylline (in co-incubation and pre-incubation conditions) for CYP1A2, 2-phenyl-2-(1-piperidinyl)propane (PPP) for CYP2B6, montelukast for CYP2C8, sulfaphenazole for CYP2C9, (-)-N-3-benzyl-phenobarbital for CYP2C19 and quinidine for CYP2D6. Quinidine 265-274 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 21131266-8 2011 Therefore, given that certain tissues (e.g., liver) express both hPXR and hCAR and that various genes are cross-regulated by them, we quantified the expression of a hCAR- and hPXR-regulated gene (CYP2B6) in cultured human hepatocytes treated with meclizine. Meclizine 247-256 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 196-202 21395541-0 2011 Steroid regulation of drug-metabolizing cytochromes P450. Steroids 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 21395541-1 2011 Cytochrome P450 (P450) monooxygenases are capable of catalyzing metabolism of various endogenous and exogenous compounds, such as bile acids, fatty acids, retinoids, steroids, drugs and other xenobiotics. Bile Acids and Salts 130-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 21395541-1 2011 Cytochrome P450 (P450) monooxygenases are capable of catalyzing metabolism of various endogenous and exogenous compounds, such as bile acids, fatty acids, retinoids, steroids, drugs and other xenobiotics. Bile Acids and Salts 130-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 21395541-1 2011 Cytochrome P450 (P450) monooxygenases are capable of catalyzing metabolism of various endogenous and exogenous compounds, such as bile acids, fatty acids, retinoids, steroids, drugs and other xenobiotics. Fatty Acids 142-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 21395541-1 2011 Cytochrome P450 (P450) monooxygenases are capable of catalyzing metabolism of various endogenous and exogenous compounds, such as bile acids, fatty acids, retinoids, steroids, drugs and other xenobiotics. Fatty Acids 142-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 21395541-1 2011 Cytochrome P450 (P450) monooxygenases are capable of catalyzing metabolism of various endogenous and exogenous compounds, such as bile acids, fatty acids, retinoids, steroids, drugs and other xenobiotics. Retinoids 155-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 21395541-1 2011 Cytochrome P450 (P450) monooxygenases are capable of catalyzing metabolism of various endogenous and exogenous compounds, such as bile acids, fatty acids, retinoids, steroids, drugs and other xenobiotics. Retinoids 155-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 21395541-1 2011 Cytochrome P450 (P450) monooxygenases are capable of catalyzing metabolism of various endogenous and exogenous compounds, such as bile acids, fatty acids, retinoids, steroids, drugs and other xenobiotics. Steroids 166-174 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 21395541-1 2011 Cytochrome P450 (P450) monooxygenases are capable of catalyzing metabolism of various endogenous and exogenous compounds, such as bile acids, fatty acids, retinoids, steroids, drugs and other xenobiotics. Steroids 166-174 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 21395541-9 2011 Modulation of P450 expression by xenobiotics can affect the subsequent metabolism of not only foreign chemicals, but also steroid hormones. Steroids 122-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 21165551-2 2011 Here, we have utilised the tumour-infiltrating properties of macrophages as vehicles to deliver a gene encoding a prodrug-activating enzyme such as human cytochrome P450 2B6 (CYP2B6) inside tumours followed by killing the tumour cells with the prodrug cyclophosphamide (CPA). Cyclophosphamide 252-268 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 175-181 21165551-2 2011 Here, we have utilised the tumour-infiltrating properties of macrophages as vehicles to deliver a gene encoding a prodrug-activating enzyme such as human cytochrome P450 2B6 (CYP2B6) inside tumours followed by killing the tumour cells with the prodrug cyclophosphamide (CPA). Cyclophosphamide 270-273 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-173 21165551-2 2011 Here, we have utilised the tumour-infiltrating properties of macrophages as vehicles to deliver a gene encoding a prodrug-activating enzyme such as human cytochrome P450 2B6 (CYP2B6) inside tumours followed by killing the tumour cells with the prodrug cyclophosphamide (CPA). Cyclophosphamide 270-273 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 175-181 21266057-6 2011 Participants responded to survey items and provided blood samples for evaluation of phenotype and genotype of CYP2A6 and CYP2B6 enzymes involved in nicotine and bupropion metabolism. Nicotine 148-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-127 21266057-6 2011 Participants responded to survey items and provided blood samples for evaluation of phenotype and genotype of CYP2A6 and CYP2B6 enzymes involved in nicotine and bupropion metabolism. Bupropion 161-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-127 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 2 subfamily B member 6 Homo sapiens 239-245 21087116-8 2011 Inhibition screening indicated that alpha-thujone inhibited both CYP2A6 and CYP2B6, with 50% inhibitory concentration values of 15.4 and 17.5 microM, respectively. beta-thujone 36-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 20825389-3 2011 CYP supersome activity for methadone use and EDDP formation ranked CYP2B6 > 3A4 > 2C19 > 2D6 > 2C18, 3A7 > 2C8, 2C9, 3A5. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 45-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 21158011-0 2011 CYP2B6 and OPRM1 gene variations predict methadone-related deaths. Methadone 41-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 21158011-3 2011 A significant association between high methadone concentrations and the CYP2B6*6 allele characteristic of the slow metabolizer phenotype was identified. Methadone 39-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 21158011-4 2011 We suggest that the risk of methadone fatality may be predetermined in part by the CYP2B6*6 allele. Methadone 28-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 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 2 subfamily B member 6 Homo sapiens 130-136 21039635-8 2011 Alcohol (100 mM) increased the mRNA levels of ABCC1 and CYP2A6 (200%), CYP2B6 and CYP3A4 (150%), and CYP2E1 (400%) compared with the control. Alcohols 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 20825389-6 2011 CYP2B6, 2D6 and 2C18 demonstrated a preference for (S)-EDDP formation; CYP2C19, 3A7 and 2C8 for (R)-EDDP; 3A4 none. (s)-eddp 51-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 20825389-6 2011 CYP2B6, 2D6 and 2C18 demonstrated a preference for (S)-EDDP formation; CYP2C19, 3A7 and 2C8 for (R)-EDDP; 3A4 none. (r)-eddp 96-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 20825389-10 2011 Chemical and mAb inhibition of CYP3A in high 3A activity HLM reduced EDDP formation by 60-85%; inhibition of CYP2B6 in 2B6 high-activity HLM reduced (S)-EDDP formation by 80% and (R)-EDDP formation by 55%. (s)-eddp 149-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 20825389-10 2011 Chemical and mAb inhibition of CYP3A in high 3A activity HLM reduced EDDP formation by 60-85%; inhibition of CYP2B6 in 2B6 high-activity HLM reduced (S)-EDDP formation by 80% and (R)-EDDP formation by 55%. (r)-eddp 179-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 20876786-0 2011 Effects of pregnane X receptor (NR1I2) and CYP2B6 genetic polymorphisms on the induction of bupropion hydroxylation by rifampin. Bupropion 92-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 21372402-1 2011 Bupropion is an atypical antidepressant that is biotransformed in humans to its major active metabolite hydroxybupropion by cytochrome P450 2B6 (CYP2B6). Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-143 21372402-1 2011 Bupropion is an atypical antidepressant that is biotransformed in humans to its major active metabolite hydroxybupropion by cytochrome P450 2B6 (CYP2B6). Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-151 21372402-1 2011 Bupropion is an atypical antidepressant that is biotransformed in humans to its major active metabolite hydroxybupropion by cytochrome P450 2B6 (CYP2B6). hydroxybupropion 104-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-143 21372402-1 2011 Bupropion is an atypical antidepressant that is biotransformed in humans to its major active metabolite hydroxybupropion by cytochrome P450 2B6 (CYP2B6). hydroxybupropion 104-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-151 21372402-2 2011 Co-administration of bupropion with an inhibitor of CYP2B6 can result in a serious drug interaction, leading to bupropion related adverse effects (e.g. seizures). Bupropion 21-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 21372402-2 2011 Co-administration of bupropion with an inhibitor of CYP2B6 can result in a serious drug interaction, leading to bupropion related adverse effects (e.g. seizures). Bupropion 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 21372402-12 2011 Our findings support future in vivo drug interaction studies in mice between bupropion and CYP2B6 inhibitors. Bupropion 77-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 20876786-0 2011 Effects of pregnane X receptor (NR1I2) and CYP2B6 genetic polymorphisms on the induction of bupropion hydroxylation by rifampin. Rifampin 119-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 20876786-3 2011 The area under the time-concentration curve (AUC) ratio of hydroxybupropion to bupropion in CYP2B6*6 carriers was significantly lower than that in CYP2B6*6 noncarriers in both the basal and rifampin-induced states (p = 0.012). hydroxybupropion 59-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 20876786-3 2011 The area under the time-concentration curve (AUC) ratio of hydroxybupropion to bupropion in CYP2B6*6 carriers was significantly lower than that in CYP2B6*6 noncarriers in both the basal and rifampin-induced states (p = 0.012). Bupropion 66-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 20876786-3 2011 The area under the time-concentration curve (AUC) ratio of hydroxybupropion to bupropion in CYP2B6*6 carriers was significantly lower than that in CYP2B6*6 noncarriers in both the basal and rifampin-induced states (p = 0.012). Rifampin 190-198 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 20876786-7 2011 In conclusion, it is suggested the NR1I2 TGT genotype decreases the bupropion hydroxylation induced by treatment with rifampin, particularly in CYP2B6*6 carriers. Bupropion 68-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-150 20876786-7 2011 In conclusion, it is suggested the NR1I2 TGT genotype decreases the bupropion hydroxylation induced by treatment with rifampin, particularly in CYP2B6*6 carriers. Rifampin 118-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-150 21869535-2 2011 In the present study, the individual differences in pharmacokinetics and/or pharmacodynamics of propofol were investigated in patients who were genotyped for CYP2B6 and UGT1A9. Propofol 96-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-164 20350955-0 2011 Effect of CYP2B6 genotype on the pharmacokinetics of sibutramine and active metabolites in healthy subjects. sibutramine 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 21869535-7 2011 The maximum plasma concentration of propofol after normalizing with the duration of infusion was affected by the CYP2B6 G516T variant (related to impaired function) and was significantly affected by a propofol risk index score that incorporated CYP2B6 G516T and UGT1A9 I399C>T (high expression) genotypes and advanced age. Propofol 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 21869535-7 2011 The maximum plasma concentration of propofol after normalizing with the duration of infusion was affected by the CYP2B6 G516T variant (related to impaired function) and was significantly affected by a propofol risk index score that incorporated CYP2B6 G516T and UGT1A9 I399C>T (high expression) genotypes and advanced age. Propofol 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 245-251 21869535-7 2011 The maximum plasma concentration of propofol after normalizing with the duration of infusion was affected by the CYP2B6 G516T variant (related to impaired function) and was significantly affected by a propofol risk index score that incorporated CYP2B6 G516T and UGT1A9 I399C>T (high expression) genotypes and advanced age. Propofol 201-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 245-251 21343660-2 2011 The (+)- and (-)-menthols were found to be oxidized to the respective (+)-(1S,3S,4S)- and (-)-(1R,3R,4R)-trans-p-menthane-3,8-diol derivatives by human liver microsomal P450 enzymes. 3-azido-2,7-naphthalene disulfonate 4-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 21343660-2 2011 The (+)- and (-)-menthols were found to be oxidized to the respective (+)-(1S,3S,4S)- and (-)-(1R,3R,4R)-trans-p-menthane-3,8-diol derivatives by human liver microsomal P450 enzymes. Menthol 13-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 21343660-2 2011 The (+)- and (-)-menthols were found to be oxidized to the respective (+)-(1S,3S,4S)- and (-)-(1R,3R,4R)-trans-p-menthane-3,8-diol derivatives by human liver microsomal P450 enzymes. (+)-(1s,3s,4s) 70-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 21343660-2 2011 The (+)- and (-)-menthols were found to be oxidized to the respective (+)-(1S,3S,4S)- and (-)-(1R,3R,4R)-trans-p-menthane-3,8-diol derivatives by human liver microsomal P450 enzymes. (-)-(1r,3r,4r)-trans-p-menthane-3,8-diol 90-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 21343660-4 2011 First, of 11 recombinant human P450 enzymes tested, CYP2A6 catalyzed the oxidation of (+)- and (-)-menthols. (+)- and (-)-menthols 86-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 21869535-0 2011 Individual differences in pharmacokinetics and pharmacodynamics of anesthetic agent propofol with regard to CYP2B6 and UGT1A9 genotype and patient age. Propofol 84-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 20350955-2 2011 The effect of CYP2B6 genotypes on the pharmacokinetics of sibutramine and its active metabolites (desmethylsibutramine [M1] and didesmethylsibutramine [M2]) was evaluated in 57 healthy subjects. sibutramine 58-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 20350955-8 2011 The CYP2B6*6 allele may be associated with a lower metabolic clearance of the M1 metabolite of sibutramine in human subjects. sibutramine 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 23226058-6 2011 When a prodrug, such as clopidogrel or codeine, must undergo hepatic biotransformation to its active form, a loss-of-function P450 genotype leads to reduced concentrations of the active drug and decreased drug efficacy. Codeine 39-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 21937854-0 2011 Biotransformation of (-)-camphor by Salmonella typhimurium OY1002/2A6 expressing human CYP2A6 and NADPH-P450 reductase. Camphor 21-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-108 21105783-0 2011 Characterization of metabolites and human P450 isoforms involved in the microsomal metabolism of mesaconitine. mesaconitine 97-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 23226058-6 2011 When a prodrug, such as clopidogrel or codeine, must undergo hepatic biotransformation to its active form, a loss-of-function P450 genotype leads to reduced concentrations of the active drug and decreased drug efficacy. Clopidogrel 24-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 21627411-4 2011 Its conversion into 2-oxo clopidogrel is regulated by cytochromes (CYP) called CYP2C19, CYP2B6 and CYP1A2. 2-oxo-clopidogrel 20-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 21105702-3 2010 The electrochemical response of the P450-electrode in the microfluidic cell was tested using four drugs that are known substrates of P450 3A4: quinidine, nifedipine, alosetron and ondansetron. Quinidine 143-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 20609441-5 2010 Norketamine was formed by CYP3A4, CYP2C19, CYP2B6, CYP2A6, CYP2D6 and CYP2C9, whereas CYP2B6 and CYP2A6 were identified to be the only enzymes which enable the hydroxylation of norketamine. norketamine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 20609441-5 2010 Norketamine was formed by CYP3A4, CYP2C19, CYP2B6, CYP2A6, CYP2D6 and CYP2C9, whereas CYP2B6 and CYP2A6 were identified to be the only enzymes which enable the hydroxylation of norketamine. norketamine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 20609441-5 2010 Norketamine was formed by CYP3A4, CYP2C19, CYP2B6, CYP2A6, CYP2D6 and CYP2C9, whereas CYP2B6 and CYP2A6 were identified to be the only enzymes which enable the hydroxylation of norketamine. norketamine 177-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 20609441-7 2010 The kinetic data of ketamine N-demethylation with CYP3A4 and CYP2B6 were best described with the Michaelis-Menten model and the Hill equation, respectively. Ketamine 20-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 21105702-3 2010 The electrochemical response of the P450-electrode in the microfluidic cell was tested using four drugs that are known substrates of P450 3A4: quinidine, nifedipine, alosetron and ondansetron. Nifedipine 154-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 21105702-3 2010 The electrochemical response of the P450-electrode in the microfluidic cell was tested using four drugs that are known substrates of P450 3A4: quinidine, nifedipine, alosetron and ondansetron. alosetron 166-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 21105702-3 2010 The electrochemical response of the P450-electrode in the microfluidic cell was tested using four drugs that are known substrates of P450 3A4: quinidine, nifedipine, alosetron and ondansetron. Ondansetron 180-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 20861742-5 2010 Darunavir, fosamprenavir, lopinavir, nelfinavir, tipranavir, efavirenz, and abacavir increased CYP3A4 and/or CYP2B6 promoter activity, some through constitutive androstane receptor but mainly through PXR. Darunavir 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 21105702-3 2010 The electrochemical response of the P450-electrode in the microfluidic cell was tested using four drugs that are known substrates of P450 3A4: quinidine, nifedipine, alosetron and ondansetron. Ondansetron 180-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 21108329-0 2010 Allelic variations in CYP2B6 and CYP2C19 and survival of patients receiving cyclophosphamide prior to myeloablative hematopoietic stem cell transplantation. Cyclophosphamide 76-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 20861742-5 2010 Darunavir, fosamprenavir, lopinavir, nelfinavir, tipranavir, efavirenz, and abacavir increased CYP3A4 and/or CYP2B6 promoter activity, some through constitutive androstane receptor but mainly through PXR. fosamprenavir 11-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 20861742-5 2010 Darunavir, fosamprenavir, lopinavir, nelfinavir, tipranavir, efavirenz, and abacavir increased CYP3A4 and/or CYP2B6 promoter activity, some through constitutive androstane receptor but mainly through PXR. Lopinavir 26-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 20861742-5 2010 Darunavir, fosamprenavir, lopinavir, nelfinavir, tipranavir, efavirenz, and abacavir increased CYP3A4 and/or CYP2B6 promoter activity, some through constitutive androstane receptor but mainly through PXR. Nelfinavir 37-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 20861742-5 2010 Darunavir, fosamprenavir, lopinavir, nelfinavir, tipranavir, efavirenz, and abacavir increased CYP3A4 and/or CYP2B6 promoter activity, some through constitutive androstane receptor but mainly through PXR. efavirenz 61-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 20861742-5 2010 Darunavir, fosamprenavir, lopinavir, nelfinavir, tipranavir, efavirenz, and abacavir increased CYP3A4 and/or CYP2B6 promoter activity, some through constitutive androstane receptor but mainly through PXR. abacavir 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 21127497-0 2010 Characterization of nuciferine metabolism by P450 enzymes and uridine diphosphate glucuronosyltransferases in liver microsomes from humans and animals. nuciferine 20-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-49 21127497-1 2010 AIM: to characterize the metabolism of nuciferine by P450 enzymes and uridine diphosphate glucuronosyltransferase (UGT) in liver microsomes from humans and several other animals including rats, mice, dogs, rabbits and monkeys. nuciferine 39-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 21127497-2 2010 METHODS: nuciferine was incubated with both human and animal liver microsomal fractions containing P450 or UGT reaction components. nuciferine 9-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-103 21127497-6 2010 RESULTS: among the nuciferine metabolites detected and identified, seven were catalyzed by P450 and one by UGT. nuciferine 19-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 21175440-7 2010 This dual genotype relationship was also observed in a preliminary clinical study, with patients who had >=1 LoF allele at either CYP2C19 or CYP2B6 also displaying significantly (P= 0.0316) lower bioactivation of cyclophosphamide. Cyclophosphamide 216-232 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-150 21175440-9 2010 CONCLUSIONS: The presence of >=1 LoF allele(s) at either CYP2B6 or CYP2C19 appeared to result in decreased bioactivation of cyclophosphamide both in vitro and in patients. Cyclophosphamide 127-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 21175440-0 2010 The combined impact of CYP2C19 and CYP2B6 pharmacogenetics on cyclophosphamide bioactivation. Cyclophosphamide 62-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 20583967-3 2010 The concentration-dependent response and time-course for the induction of CYP1A2, CYP2B6 and CYP3A4 by inducing agents beta-naphthoflavone, phenobarbital and rifampicin, respectively were examined in two or more donors using multiple end-points (mRNA, enzyme activity and Western blot analysis). beta-Naphthoflavone 119-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 20583967-3 2010 The concentration-dependent response and time-course for the induction of CYP1A2, CYP2B6 and CYP3A4 by inducing agents beta-naphthoflavone, phenobarbital and rifampicin, respectively were examined in two or more donors using multiple end-points (mRNA, enzyme activity and Western blot analysis). Phenobarbital 140-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 20583967-3 2010 The concentration-dependent response and time-course for the induction of CYP1A2, CYP2B6 and CYP3A4 by inducing agents beta-naphthoflavone, phenobarbital and rifampicin, respectively were examined in two or more donors using multiple end-points (mRNA, enzyme activity and Western blot analysis). Rifampin 158-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 20736323-6 2010 The inhibitory potency of DBZ toward CYP3A4 was greater than that toward other P450 isoforms, including CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2D6. dibenzazepine 26-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 20736323-1 2010 Nonsynonymous single-nucleotide polymorphisms (nsSNPs) in cytochrome P450 (P450) genes may affect drug metabolism and drug-drug interactions (DDIs), potentially leading to adverse drug reactions. single-nucleotide 14-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-73 20736323-1 2010 Nonsynonymous single-nucleotide polymorphisms (nsSNPs) in cytochrome P450 (P450) genes may affect drug metabolism and drug-drug interactions (DDIs), potentially leading to adverse drug reactions. single-nucleotide 14-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 20736323-8 2010 These results provide useful information for understanding the influence of P450 genetic polymorphisms on DBZ metabolism and may help to design future clinical trials of DBZ. dibenzazepine 106-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 20736323-8 2010 These results provide useful information for understanding the influence of P450 genetic polymorphisms on DBZ metabolism and may help to design future clinical trials of DBZ. dibenzazepine 170-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 20736323-3 2010 To evaluate the effects of P450 nsSNPs on the metabolism and inhibition potential of a candidate drug, tanshinol borneol ester (DBZ), we obtained and experimentally validated eight yeast-expressed human P450 isoforms and their nsSNP variants and tested DBZ using these recombinant P450 enzymes. 1,7,7-trimethylbicyclo(2.2.1)heptan-2-yl 3-(3,4-dihydroxyphenyl)-2-hydroxypropanoate 103-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 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 2 subfamily B member 6 Homo sapiens 36-40 20736323-3 2010 To evaluate the effects of P450 nsSNPs on the metabolism and inhibition potential of a candidate drug, tanshinol borneol ester (DBZ), we obtained and experimentally validated eight yeast-expressed human P450 isoforms and their nsSNP variants and tested DBZ using these recombinant P450 enzymes. 1,7,7-trimethylbicyclo(2.2.1)heptan-2-yl 3-(3,4-dihydroxyphenyl)-2-hydroxypropanoate 103-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 203-207 20736323-3 2010 To evaluate the effects of P450 nsSNPs on the metabolism and inhibition potential of a candidate drug, tanshinol borneol ester (DBZ), we obtained and experimentally validated eight yeast-expressed human P450 isoforms and their nsSNP variants and tested DBZ using these recombinant P450 enzymes. 1,7,7-trimethylbicyclo(2.2.1)heptan-2-yl 3-(3,4-dihydroxyphenyl)-2-hydroxypropanoate 103-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 203-207 20736323-3 2010 To evaluate the effects of P450 nsSNPs on the metabolism and inhibition potential of a candidate drug, tanshinol borneol ester (DBZ), we obtained and experimentally validated eight yeast-expressed human P450 isoforms and their nsSNP variants and tested DBZ using these recombinant P450 enzymes. dibenzazepine 128-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 203-207 20736323-3 2010 To evaluate the effects of P450 nsSNPs on the metabolism and inhibition potential of a candidate drug, tanshinol borneol ester (DBZ), we obtained and experimentally validated eight yeast-expressed human P450 isoforms and their nsSNP variants and tested DBZ using these recombinant P450 enzymes. dibenzazepine 128-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 203-207 20851877-5 2010 A good correlation was observed between sesamin catecholization activity and CYP2C9-specific activity in in vitro studies using 10 individual human liver microsomes, strongly suggesting that CYP2C9 is the most important P450 isoform for sesamin catecholization in human liver. sesamin 40-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-224 20851877-4 2010 Based on the kinetic data and average contents of the P450 isoforms in the human liver, the putative contribution of P450s for sesamin metabolism was large in the order of CYP2C9, 1A2, 2C19, and 2D6. sesamin 127-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 20851877-5 2010 A good correlation was observed between sesamin catecholization activity and CYP2C9-specific activity in in vitro studies using 10 individual human liver microsomes, strongly suggesting that CYP2C9 is the most important P450 isoform for sesamin catecholization in human liver. sesamin 237-244 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-224 20863199-11 2010 Although pre-incubation did enhance the potency for CYP2B6 inhibition to 5.1 muM, given that exposure to capsaicin from either food or a topical medicine is very low (<=58 nM) and transient, effects on CYPs appear unlikely. Capsaicin 105-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 20851877-7 2010 We also examined the inhibitory effect of sesamin for P450 isoform-specific activities and found a mechanism-based inhibition of CYP2C9 by sesamin. sesamin 42-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 21053990-19 2010 Prasugrel did not affect the activities of CYP2C9, CYP2C19 or P-glycoprotein, but it weakly inhibited CYP2B6. Prasugrel Hydrochloride 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 20829393-4 2010 In HepG2 cells, buprenorphine significantly increased human PXR-mediated CYP2B6 and CYP3A4 reporter activities. Buprenorphine 16-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 20829393-5 2010 CYP2B6 reporter activity was also enhanced by buprenorphine in HepG2 cells cotransfected with a chemical-responsive human CAR variant. Buprenorphine 46-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 20849814-1 2010 Cytochrome P450 3A4 (CYP3A4), the major P450 present in human liver metabolizes approximately half the drugs in clinical use and requires electrons supplied from NADPH through NADPH-P450 reductase (POR, CPR). NADP 162-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 21918647-1 2010 The human drug metabolizing cytochrome P450 (CYP) 1A2, is one of the major P450 isoforms contributing by about 5-20% to the hepatic P450 pool and catalyzing oxidative biotransformation of up to 10% of clinically relevant drugs including clozapine and caffeine. Clozapine 237-246 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 21918647-1 2010 The human drug metabolizing cytochrome P450 (CYP) 1A2, is one of the major P450 isoforms contributing by about 5-20% to the hepatic P450 pool and catalyzing oxidative biotransformation of up to 10% of clinically relevant drugs including clozapine and caffeine. Clozapine 237-246 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 21918647-1 2010 The human drug metabolizing cytochrome P450 (CYP) 1A2, is one of the major P450 isoforms contributing by about 5-20% to the hepatic P450 pool and catalyzing oxidative biotransformation of up to 10% of clinically relevant drugs including clozapine and caffeine. Caffeine 251-259 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 21918647-1 2010 The human drug metabolizing cytochrome P450 (CYP) 1A2, is one of the major P450 isoforms contributing by about 5-20% to the hepatic P450 pool and catalyzing oxidative biotransformation of up to 10% of clinically relevant drugs including clozapine and caffeine. Caffeine 251-259 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 20709133-6 2010 Chlorpyrifos metabolism by individual human liver microsomes was significantly correlated with CYP2B6, CYP2C19 and CYP3A4 related activity. Chlorpyrifos 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-101 20709133-7 2010 CPO formation was best correlated with CYP2B6 related activity at low (20 muM) chlorpyrifos concentrations while CYP3A4 related activity was best correlated with CPO formation at high concentrations (100 muM) of chlorpyrifos. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 20709133-7 2010 CPO formation was best correlated with CYP2B6 related activity at low (20 muM) chlorpyrifos concentrations while CYP3A4 related activity was best correlated with CPO formation at high concentrations (100 muM) of chlorpyrifos. Chlorpyrifos 79-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 20849814-1 2010 Cytochrome P450 3A4 (CYP3A4), the major P450 present in human liver metabolizes approximately half the drugs in clinical use and requires electrons supplied from NADPH through NADPH-P450 reductase (POR, CPR). NADP 162-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-44 20668445-0 2010 OPRM1 and CYP2B6 gene variants as risk factors in methadone-related deaths. Methadone 50-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 21103422-2 2010 One macrolide, erythromycin (ERY), is associated with possible sudden cardiac death from QT prolongation due to P450 iso-enzyme inhibition. Macrolides 4-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 21103422-2 2010 One macrolide, erythromycin (ERY), is associated with possible sudden cardiac death from QT prolongation due to P450 iso-enzyme inhibition. Erythromycin 15-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 21103422-2 2010 One macrolide, erythromycin (ERY), is associated with possible sudden cardiac death from QT prolongation due to P450 iso-enzyme inhibition. Erythromycin 29-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 21103422-3 2010 An alternative, azithromycin (AZI), lacks P450 inhibition. Azithromycin 16-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 20836875-0 2010 Adenoviral delivery of pan-caspase inhibitor p35 enhances bystander killing by P450 gene-directed enzyme prodrug therapy using cyclophosphamide+. Cyclophosphamide 127-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 20836875-1 2010 BACKGROUND: Cytochrome P450-based suicide gene therapy for cancer using prodrugs such as cyclophosphamide (CPA) increases anti-tumor activity, both directly and via a bystander killing mechanism. Cyclophosphamide 89-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 20836875-1 2010 BACKGROUND: Cytochrome P450-based suicide gene therapy for cancer using prodrugs such as cyclophosphamide (CPA) increases anti-tumor activity, both directly and via a bystander killing mechanism. Cyclophosphamide 107-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 20836875-3 2010 Previous studies have shown that the pan-caspase inhibitor p35 significantly increases CPA-induced bystander killing by tumor cells that stably express P450 enzyme CYP2B6 (Schwartz et al, (2002) Cancer Res. Cyclophosphamide 87-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-170 20709133-10 2010 Calculations of percent total normalized rates (% TNR) and the chemical inhibitors ketoconazole and ticlopidine were used to confirm the importance of CYP2B6, CYP2C19, and CYP3A4 for the metabolism of chlorpyrifos. Chlorpyrifos 201-213 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 20624854-5 2010 Cell-based promoter reporter assays demonstrated that CYP2B6 luciferase activity was synergistically enhanced in the presence of both -82T C mutation and rifampicin (RIF)-activated PXR. Rifampin 154-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 20624854-5 2010 Cell-based promoter reporter assays demonstrated that CYP2B6 luciferase activity was synergistically enhanced in the presence of both -82T C mutation and rifampicin (RIF)-activated PXR. Rifampin 166-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 20735006-4 2010 Here we show that electrochemical generation of reactive oxygen species (ROS), a process reminiscent of the catalytic cycle of P450, extends the applicability of electrochemistry in drug metabolism research. Reactive Oxygen Species 48-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 20735006-4 2010 Here we show that electrochemical generation of reactive oxygen species (ROS), a process reminiscent of the catalytic cycle of P450, extends the applicability of electrochemistry in drug metabolism research. Reactive Oxygen Species 73-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 20668445-8 2010 The risk of a methadone-related fatality during treatment may be evaluated in part by screening for CYP2B6*6 and A118G. Methadone 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 20668445-2 2010 We have examined the association between CYP2B6 and micro-opioid receptor (OPRM1) gene variations and apparent susceptibility to methadone poisoning. Methadone 129-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 20668445-5 2010 CYP2B6 *4, *9, and *6 alleles were found to be associated with higher postmortem methadone concentrations in blood (P < or = 0.05). Methadone 81-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 20885377-1 2010 Mechanism-based inactivators such as bergamottin are useful chemical tools for identifying the functions of specific active-site amino acid residues in the reactions catalyzed by cytochromes P450 (CYPs or P450s), which are responsible for the metabolism of a wide variety of drugs and endogenous substrates. bergamottin 37-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 191-195 20516253-5 2010 The expression of CYP2B6 mRNA was induced in HepG2 cells stably expressing mouse CAR (Ym17) by HDAC inhibitors including valproic acid, phenylbutyrate, and trichostatin A. Valproic Acid 121-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 20516253-5 2010 The expression of CYP2B6 mRNA was induced in HepG2 cells stably expressing mouse CAR (Ym17) by HDAC inhibitors including valproic acid, phenylbutyrate, and trichostatin A. Phenylbutyrates 136-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 20516253-5 2010 The expression of CYP2B6 mRNA was induced in HepG2 cells stably expressing mouse CAR (Ym17) by HDAC inhibitors including valproic acid, phenylbutyrate, and trichostatin A. trichostatin A 156-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 20516253-6 2010 HDAC inhibitors activated the phenobarbital-responsive enhancer module of the CYP2B6 promoter in transient transfection reporter assays with Ym17 cells. Phenobarbital 30-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 20516253-7 2010 Furthermore, HDAC inhibitors synergistically augmented the effect of the CAR ligand, 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene, in the transactivation of CYP2B6 mRNA and the promoter assay in Ym17 cells. 1,4-bis(2-(3,5-dichloropyridyloxy))benzene 85-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-161 20501634-0 2010 The effects of azole-based heme oxygenase inhibitors on rat cytochromes P450 2E1 and 3A1/2 and human cytochromes P450 3A4 and 2D6. Azoles 15-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-76 20501634-4 2010 However, these tools are limited by their lack of selectivity; they affect other heme-dependent enzymes, such as cytochromes P450 (P450s), soluble guanylyl cyclase (sGC), and nitric-oxide synthase (NOS). Heme 81-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-129 20501634-4 2010 However, these tools are limited by their lack of selectivity; they affect other heme-dependent enzymes, such as cytochromes P450 (P450s), soluble guanylyl cyclase (sGC), and nitric-oxide synthase (NOS). Heme 81-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-136 20885377-1 2010 Mechanism-based inactivators such as bergamottin are useful chemical tools for identifying the functions of specific active-site amino acid residues in the reactions catalyzed by cytochromes P450 (CYPs or P450s), which are responsible for the metabolism of a wide variety of drugs and endogenous substrates. bergamottin 37-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 205-210 20860463-12 2010 CONCLUSION: Our study corroborates previous findings indicating that knowledge of CYP2B6 genetic status should be taken into account for an EFV treatment. efavirenz 140-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 20669986-1 2010 The in vitro bioactivation of the selective serotonin and norepinephrine reuptake inhibitor duloxetine was investigated using liver microsomes and cytosol, expressed glutathione transferase, and recombinant P450 2D6 and 1A2. Duloxetine Hydrochloride 92-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 207-211 20669986-4 2010 Experiments with recombinant P450s and the isoform specific inhibitors quinidine and furafylline suggested that both P450 2D6 and 1A2 were involved in the bioactivation of duloxetine. Duloxetine Hydrochloride 172-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 20707407-0 2010 Cloning, expression and functional characterization of cytochrome P450 3A37 from turkey liver with high aflatoxin B1 epoxidation activity. Aflatoxins 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 20669986-6 2010 MetaSite and induced fit docking but not rigid docking correctly predicted that naphthalene rather than thiophene was the preferred site of bioactivation for duloxetine by P450 2D6. naphthalene 80-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 20707407-3 2010 Poultry, especially turkeys are extremely sensitive to AFB(1), a condition due, in part, to efficient epoxidation by P450 1A and 3A enzymes. Aflatoxin B1 55-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-121 20669986-6 2010 MetaSite and induced fit docking but not rigid docking correctly predicted that naphthalene rather than thiophene was the preferred site of bioactivation for duloxetine by P450 2D6. Thiophenes 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 20707407-8 2010 Like human P450 3A4, 3A37 biotransformed AFB(1) to exo-AFBO and aflatoxin Q(1) (AFQ(1)) and possessed nifedipine oxidation activity, both of which were inhibited by the P450 3A4 inhibitor 17alpha-ethynylestradiol. aflatoxin Q1 64-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 20707407-8 2010 Like human P450 3A4, 3A37 biotransformed AFB(1) to exo-AFBO and aflatoxin Q(1) (AFQ(1)) and possessed nifedipine oxidation activity, both of which were inhibited by the P450 3A4 inhibitor 17alpha-ethynylestradiol. aflatoxin Q1 64-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 20669986-6 2010 MetaSite and induced fit docking but not rigid docking correctly predicted that naphthalene rather than thiophene was the preferred site of bioactivation for duloxetine by P450 2D6. Duloxetine Hydrochloride 158-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 20335270-5 2010 Both 7- and 8-hydroxyefavirenz were further oxidized to novel dihydroxylated metabolite(s) primarily by CYP2B6. 7- and 8-hydroxyefavirenz 5-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-110 20707407-8 2010 Like human P450 3A4, 3A37 biotransformed AFB(1) to exo-AFBO and aflatoxin Q(1) (AFQ(1)) and possessed nifedipine oxidation activity, both of which were inhibited by the P450 3A4 inhibitor 17alpha-ethynylestradiol. 6-Amino-4-{2-[(Cyclopentylmethyl)amino]ethyl}-2-(Methylamino)-1,7-Dihydro-8h-Imidazo[4,5-G]quinazolin-8-One 80-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 20707407-8 2010 Like human P450 3A4, 3A37 biotransformed AFB(1) to exo-AFBO and aflatoxin Q(1) (AFQ(1)) and possessed nifedipine oxidation activity, both of which were inhibited by the P450 3A4 inhibitor 17alpha-ethynylestradiol. Nifedipine 102-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 20707407-8 2010 Like human P450 3A4, 3A37 biotransformed AFB(1) to exo-AFBO and aflatoxin Q(1) (AFQ(1)) and possessed nifedipine oxidation activity, both of which were inhibited by the P450 3A4 inhibitor 17alpha-ethynylestradiol. Nifedipine 102-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 20707407-8 2010 Like human P450 3A4, 3A37 biotransformed AFB(1) to exo-AFBO and aflatoxin Q(1) (AFQ(1)) and possessed nifedipine oxidation activity, both of which were inhibited by the P450 3A4 inhibitor 17alpha-ethynylestradiol. Ethinyl Estradiol 188-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 20707407-8 2010 Like human P450 3A4, 3A37 biotransformed AFB(1) to exo-AFBO and aflatoxin Q(1) (AFQ(1)) and possessed nifedipine oxidation activity, both of which were inhibited by the P450 3A4 inhibitor 17alpha-ethynylestradiol. Ethinyl Estradiol 188-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 20707407-12 2010 These data strongly suggest that P450 3A37, along with P450 1A5, plays an important role in AFB(1) epoxidation in turkey liver. Aflatoxin B1 92-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 20707407-12 2010 These data strongly suggest that P450 3A37, along with P450 1A5, plays an important role in AFB(1) epoxidation in turkey liver. Aflatoxin B1 92-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 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 2 subfamily B member 6 Homo sapiens 151-157 20642445-0 2010 Induction of CYP2B6 and CYP3A4 expression by 1-aminobenzotriazole (ABT) in human hepatocytes. 1-aminobenzotriazole 45-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 20642445-0 2010 Induction of CYP2B6 and CYP3A4 expression by 1-aminobenzotriazole (ABT) in human hepatocytes. 1-aminobenzotriazole 67-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 20642445-3 2010 In this study, we show that ABT up-regulates expression of CYP2B6 and CYP3A4 potentially by activating nuclear receptor CAR. 1-aminobenzotriazole 28-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 20642445-4 2010 In freshly isolated human hepatocytes, ABT increased mRNA expression of CYP2B6 and CYP3A4 in a concentration-dependent manner. 1-aminobenzotriazole 39-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 20642445-5 2010 ABT also modulated CYP-inducing actions of CITCO and rifampin, the known inducers of CYP2B6 and CYP3A4. 1-aminobenzotriazole 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20642445-5 2010 ABT also modulated CYP-inducing actions of CITCO and rifampin, the known inducers of CYP2B6 and CYP3A4. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 43-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20642445-5 2010 ABT also modulated CYP-inducing actions of CITCO and rifampin, the known inducers of CYP2B6 and CYP3A4. Rifampin 53-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20642445-6 2010 Results from luciferase reporter assays confirmed that ABT increases CYP2B6 promoter activity in CAR-expressing HepG2 cells. 1-aminobenzotriazole 55-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 20509749-3 2010 In liver microsomes from PB-treated pigs, these transcriptional activations were accompanied by an increase of various marker activities of human CYP2B6, CYP3As, CYP2C9, CYP2C19. Phenobarbital 25-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 146-152 20528624-1 2010 The involvement of cytochrome P450 2B6 (CYP2B6) to the in vitro and in vivo metabolism of bupropion has been well studied. Bupropion 90-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-38 20528624-1 2010 The involvement of cytochrome P450 2B6 (CYP2B6) to the in vitro and in vivo metabolism of bupropion has been well studied. Bupropion 90-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 20528624-4 2010 Incubations in rhCYP identified CYP2B6 as the isoform responsible for the formation of hydroxybupropion (M3). hydroxybupropion 87-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 20528624-8 2010 These results confirmed the principle role of CYP2B6 in hydroxybupropion formation, as a selective CYP2B6 probe. hydroxybupropion 56-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 20528624-8 2010 These results confirmed the principle role of CYP2B6 in hydroxybupropion formation, as a selective CYP2B6 probe. hydroxybupropion 56-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 20642555-3 2010 Two terpenoids, borneol and isoborneol, are major constituents of woohwangcheongsimwon suspension, and show a competitive inhibition of CYP2B6 with K(i) values of 9.5 and 5.9 microM, respectively. Terpenes 4-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 20642555-3 2010 Two terpenoids, borneol and isoborneol, are major constituents of woohwangcheongsimwon suspension, and show a competitive inhibition of CYP2B6 with K(i) values of 9.5 and 5.9 microM, respectively. isoborneol 16-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 20642555-3 2010 Two terpenoids, borneol and isoborneol, are major constituents of woohwangcheongsimwon suspension, and show a competitive inhibition of CYP2B6 with K(i) values of 9.5 and 5.9 microM, respectively. isoborneol 28-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 20642555-4 2010 Bupropion undergoes metabolic transformation to the active metabolite, 4-hydroxybupropion, primarily via CYP2B6 both in vivo and in vitro. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 20642555-4 2010 Bupropion undergoes metabolic transformation to the active metabolite, 4-hydroxybupropion, primarily via CYP2B6 both in vivo and in vitro. hydroxybupropion 71-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 20642555-9 2010 AIMS: To examine the effects of woohwangcheongsimwon suspension on the pharmacokinetics of bupropion and its active metabolite, 4-hydroxybupropion, formed via CYP2B6 in vivo. Bupropion 91-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 20642555-9 2010 AIMS: To examine the effects of woohwangcheongsimwon suspension on the pharmacokinetics of bupropion and its active metabolite, 4-hydroxybupropion, formed via CYP2B6 in vivo. hydroxybupropion 128-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 20361990-0 2010 Endosulfan induces CYP2B6 and CYP3A4 by activating the pregnane X receptor. Endosulfan 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 20361990-11 2010 These data support the role of endosulfan-alpha as a strong activator of PXR and inducer of CYP2B6 and CYP3A4, which may impact metabolism of CYP2B6 or CYP3A4 substrates. Endosulfan 31-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 20361990-11 2010 These data support the role of endosulfan-alpha as a strong activator of PXR and inducer of CYP2B6 and CYP3A4, which may impact metabolism of CYP2B6 or CYP3A4 substrates. Endosulfan 31-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 20089352-0 2010 The influence of cytochrome oxidase CYP2A6, CYP2B6, and CYP2C9 polymorphisms on the plasma concentrations of valproic acid in epileptic patients. Valproic Acid 109-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 20109440-8 2010 On the other hand, CYP2B6 was responsible for the formation of OH-bupropion. oh-bupropion 63-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 20307138-6 2010 Female livers and livers exposed to inducers (phenobarbital and/or dexamethasone) were associated with higher CYP2B6. Phenobarbital 46-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 20307138-6 2010 Female livers and livers exposed to inducers (phenobarbital and/or dexamethasone) were associated with higher CYP2B6. Dexamethasone 67-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 20307138-7 2010 A weak correlation between CYP2B6 and nicotine C-oxidation activity was observed, which was abrogated when controlling for CYP2A6 protein levels. Nicotine 38-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 20307514-7 2010 CYP3A5 and CYP2B6 had the greatest efficiency to form carbosulfan sulfinamide, while CYP3A4 and CYP3A5 were the most efficient in the generation of the carbofuran metabolic pathway. carbosulfan sulfinamide 54-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 20307514-7 2010 CYP3A5 and CYP2B6 had the greatest efficiency to form carbosulfan sulfinamide, while CYP3A4 and CYP3A5 were the most efficient in the generation of the carbofuran metabolic pathway. Carbofuran 152-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 20100815-0 2010 Engineering bacterial cytochrome P450 (P450) BM3 into a prototype with human P450 enzyme activity using indigo formation. Indigo Carmine 104-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-43 20100815-6 2010 When the P450 activities were measured at the whole-cell level, some of the blue colonies, but not the white colonies, possessed apparent oxidation activity toward coumarin and 7-ethoxycoumarin, which are typical human P450 substrates that produce fluorescent products. 7-ethoxycoumarin 177-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 219-223 20100815-0 2010 Engineering bacterial cytochrome P450 (P450) BM3 into a prototype with human P450 enzyme activity using indigo formation. Indigo Carmine 104-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 20100815-9 2010 Highly active mutants are also able to metabolize several other human P450 substrates, including phenacetin, ethoxyresorufin, and chlorzoxazone. Phenacetin 97-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 20100815-9 2010 Highly active mutants are also able to metabolize several other human P450 substrates, including phenacetin, ethoxyresorufin, and chlorzoxazone. ethoxyresorufin 109-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 20100815-1 2010 Human cytochrome P450 (P450) enzymes metabolize a variety of endogenous and xenobiotic compounds, including steroids, drugs, and environmental chemicals. Steroids 108-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-27 20100815-9 2010 Highly active mutants are also able to metabolize several other human P450 substrates, including phenacetin, ethoxyresorufin, and chlorzoxazone. Chlorzoxazone 130-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 20100815-6 2010 When the P450 activities were measured at the whole-cell level, some of the blue colonies, but not the white colonies, possessed apparent oxidation activity toward coumarin and 7-ethoxycoumarin, which are typical human P450 substrates that produce fluorescent products. coumarin 164-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-13 20100815-10 2010 These results indicate that indigo formation provides a simple assay for identifying CYP102A1 mutants with a greater potential for human P450 activity. Indigo Carmine 28-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-141 20100815-6 2010 When the P450 activities were measured at the whole-cell level, some of the blue colonies, but not the white colonies, possessed apparent oxidation activity toward coumarin and 7-ethoxycoumarin, which are typical human P450 substrates that produce fluorescent products. coumarin 164-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 219-223 20100815-6 2010 When the P450 activities were measured at the whole-cell level, some of the blue colonies, but not the white colonies, possessed apparent oxidation activity toward coumarin and 7-ethoxycoumarin, which are typical human P450 substrates that produce fluorescent products. 7-ethoxycoumarin 177-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-13 20338069-1 2010 BACKGROUND: Cytochrome P450 2B6 (CYP2B6) metabolizes efavirenz and nevirapine, the major core antiretroviral drugs for HIV in Thailand. efavirenz 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-31 20061448-1 2010 The structure of the K262R genetic variant of human cytochrome P450 2B6 in complex with the inhibitor 4-(4-chlorophenyl)imidazole (4-CPI) has been determined using X-ray crystallography to 2.0-A resolution. 4-(4-chlorophenyl)imidazole 131-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-71 20148932-14 2010 Our results help to show the diversity of P450 and alkB genes in prokaryotes, and to portray the geographic distribution of oil-degrading bacteria in marine environments. Oils 124-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-55 20149798-6 2010 The access channel is typical for mammalian sterol-metabolizing P450 enzymes, but is different from that observed in Mycobacterium tuberculosis CYP51. Sterols 44-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 20149798-7 2010 Comparison of the azole-bound structures provides insight into the relative binding affinities of human and bacterial P450 enzymes to ketoconazole and fluconazole, which can be useful for the rational design of antifungal compounds and specific modulators of human CYP51. Azoles 18-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 20149798-7 2010 Comparison of the azole-bound structures provides insight into the relative binding affinities of human and bacterial P450 enzymes to ketoconazole and fluconazole, which can be useful for the rational design of antifungal compounds and specific modulators of human CYP51. Ketoconazole 134-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 20149798-7 2010 Comparison of the azole-bound structures provides insight into the relative binding affinities of human and bacterial P450 enzymes to ketoconazole and fluconazole, which can be useful for the rational design of antifungal compounds and specific modulators of human CYP51. Fluconazole 151-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 20061448-0 2010 Crystal structure of a cytochrome P450 2B6 genetic variant in complex with the inhibitor 4-(4-chlorophenyl)imidazole at 2.0-A resolution. 4-(4-chlorophenyl)imidazole 89-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-42 20061448-1 2010 The structure of the K262R genetic variant of human cytochrome P450 2B6 in complex with the inhibitor 4-(4-chlorophenyl)imidazole (4-CPI) has been determined using X-ray crystallography to 2.0-A resolution. 4-(4-chlorophenyl)imidazole 102-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-71 20338069-1 2010 BACKGROUND: Cytochrome P450 2B6 (CYP2B6) metabolizes efavirenz and nevirapine, the major core antiretroviral drugs for HIV in Thailand. efavirenz 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 20338069-1 2010 BACKGROUND: Cytochrome P450 2B6 (CYP2B6) metabolizes efavirenz and nevirapine, the major core antiretroviral drugs for HIV in Thailand. Nevirapine 67-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-31 20338069-1 2010 BACKGROUND: Cytochrome P450 2B6 (CYP2B6) metabolizes efavirenz and nevirapine, the major core antiretroviral drugs for HIV in Thailand. Nevirapine 67-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 20338069-6 2010 The frequencies of GG, GT and TT genotypes of CYP2B6-G516T were 38.46%, 47.69% and 13.85% in efavirenz group and 44.07%, 52.54% and 3.39% in nevirapine group, respectively. Nevirapine 141-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 20338069-11 2010 This is the first report to demonstrate the effects of CYP2B6 G516T polymorphisms on plasma efavirenz and nevirapine concentrations when co-administered with rifampicin in HIV/TB co-infected Thai adults. Rifampin 158-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 20338069-12 2010 CONCLUSIONS: CYP2B6-TT genotype had impact on plasma efavirenz and nevirapine concentrations, while rifampicin co-administration had only small effects. Nevirapine 67-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 20068028-7 2010 A peculiar P450, P450nor, receives electrons directly from NADH for the reduction of nitric oxide. NAD 59-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 20102294-1 2010 The objective of this study was to investigate the effects of continuous St. John"s wort administration on single-dose pharmacokinetics of bupropion, a substrate of cytochrome P450 (CYP) 2B6, in healthy Chinese volunteers. Bupropion 139-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-190 20019244-2 2010 In the present study, we carefully analyzed mRNA expression and activity of the major P450s and their responsiveness to three prototypical inducers, phenobarbital, rifampicin, and omeprazole, in differentiated HepaRG cell cultures over a 4-week period after low and high seeding. Phenobarbital 149-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-91 20019244-2 2010 In the present study, we carefully analyzed mRNA expression and activity of the major P450s and their responsiveness to three prototypical inducers, phenobarbital, rifampicin, and omeprazole, in differentiated HepaRG cell cultures over a 4-week period after low and high seeding. Rifampin 164-174 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-91 20019244-2 2010 In the present study, we carefully analyzed mRNA expression and activity of the major P450s and their responsiveness to three prototypical inducers, phenobarbital, rifampicin, and omeprazole, in differentiated HepaRG cell cultures over a 4-week period after low and high seeding. Omeprazole 180-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-91 20019244-5 2010 Expression and activities of several P450s were dimethyl sulfoxide-dependent. Dimethyl Sulfoxide 48-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-42 20019244-7 2010 Thus, CYP1A2, CYP2B6, and CYP3A4 were found to accurately respond to their respective prototypical inducers, i.e., omeprazole, phenobarbital, and rifampicin. Omeprazole 115-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 20019244-7 2010 Thus, CYP1A2, CYP2B6, and CYP3A4 were found to accurately respond to their respective prototypical inducers, i.e., omeprazole, phenobarbital, and rifampicin. Phenobarbital 127-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 20019244-7 2010 Thus, CYP1A2, CYP2B6, and CYP3A4 were found to accurately respond to their respective prototypical inducers, i.e., omeprazole, phenobarbital, and rifampicin. Rifampin 146-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 19653275-5 2010 NSPC were then coinjected into mouse brain with GBM cells, employing NSPC expressing cyclophosphamide (CPA)-activating enzyme cytochrome p450 2B6 (CYP2B6), which catalyzes CPA prodrug transformation into membrane diffusible DNA-alkylating metabolites. Cyclophosphamide 85-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 19653275-5 2010 NSPC were then coinjected into mouse brain with GBM cells, employing NSPC expressing cyclophosphamide (CPA)-activating enzyme cytochrome p450 2B6 (CYP2B6), which catalyzes CPA prodrug transformation into membrane diffusible DNA-alkylating metabolites. Cyclophosphamide 103-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 19653275-5 2010 NSPC were then coinjected into mouse brain with GBM cells, employing NSPC expressing cyclophosphamide (CPA)-activating enzyme cytochrome p450 2B6 (CYP2B6), which catalyzes CPA prodrug transformation into membrane diffusible DNA-alkylating metabolites. Cyclophosphamide 172-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 19653275-6 2010 Upon CPA administration, NSPC containing CYP2B6 elicited substantial impairment of tumor growth. Cyclophosphamide 5-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 20068028-7 2010 A peculiar P450, P450nor, receives electrons directly from NADH for the reduction of nitric oxide. Nitric Oxide 85-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 19833192-0 2010 Involvement of pregnane X receptor in the regulation of CYP2B6 gene expression by oltipraz in human hepatocytes. oltipraz 82-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 19833192-10 2010 This can be explained by a dose-dependent inhibition of CYP2B6 activity in presence of oltipraz as demonstrated with human hepatocyte microsomes. oltipraz 87-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 19833192-2 2010 Previously, we have demonstrated that CYP2B6 expression is induced in cultured human hepatocytes by a 24h treatment with oltipraz. oltipraz 121-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 19833192-11 2010 Altogether, this study provides the first demonstration of PXR involvement in oltipraz transcriptional activation of CYP2B6 gene and of the inhibitory effect of oltipraz on CYP2B6 activity. oltipraz 78-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 19833192-11 2010 Altogether, this study provides the first demonstration of PXR involvement in oltipraz transcriptional activation of CYP2B6 gene and of the inhibitory effect of oltipraz on CYP2B6 activity. oltipraz 161-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 19833192-5 2010 The rapid induction of CYP2B6 mRNA in oltipraz-treated cells suggests a transcriptional activation of corresponding gene. oltipraz 38-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 19833192-7 2010 The results demonstrate that transcriptional activation of CYP2B6 gene is mediated mainly by the pregnane X receptor (PXR) and the Phenobarbital Responsive Element Module (PBREM). Phenobarbital 131-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 20610889-0 2010 Enhanced susceptibility of HLA-mediated ticlopidine-induced idiosyncratic hepatotoxicity by CYP2B6 polymorphism in Japanese. Ticlopidine 40-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 19944064-5 2010 Compressibility of the heme pocket in variants of all four CYP2B enzymes containing proline at position 334 are characterized by lower compressibility than their more stable serine 334 counterpart. Heme 23-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-64 19944064-5 2010 Compressibility of the heme pocket in variants of all four CYP2B enzymes containing proline at position 334 are characterized by lower compressibility than their more stable serine 334 counterpart. Proline 84-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-64 19944064-5 2010 Compressibility of the heme pocket in variants of all four CYP2B enzymes containing proline at position 334 are characterized by lower compressibility than their more stable serine 334 counterpart. Serine 174-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-64 19797611-2 2010 Treatment with 1,4-diamino-2,3-dicyano-1,4-bis(methylthio)butadiene (U0126) (an extracellular signal-regulated kinase inhibitor) suppressed the HGF-induced expression of UGT1A1 and CYP2B6, as well as p16, p21, and p27 in HepG2 cells. u0124 15-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-187 19797611-2 2010 Treatment with 1,4-diamino-2,3-dicyano-1,4-bis(methylthio)butadiene (U0126) (an extracellular signal-regulated kinase inhibitor) suppressed the HGF-induced expression of UGT1A1 and CYP2B6, as well as p16, p21, and p27 in HepG2 cells. U 0126 69-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-187 19797611-3 2010 The CDK inhibitor roscovitine also enhanced the expression of UGT1A1, CYP2B6, and CYP3A4. Roscovitine 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 19497360-4 2010 In contrast, the fold-induction of CYP2B6 by PB was 3-fold higher that by 10microM RIF. Lead 45-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 19797611-6 2010 In cells arrested in S phase by a thymidine block and then released into a synchronous cell cycle, there was a clear dissociation among the activation of CDK2 and the expression of UGT1A1, CYP2B6, and CYP3A4. Thymidine 34-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-195 19812348-0 2010 Identification of the human cytochrome P450 enzymes involved in the two oxidative steps in the bioactivation of clopidogrel to its pharmacologically active metabolite. Clopidogrel 112-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 19812348-1 2010 The aim of the current study is to identify the human cytochrome P450 (P450) isoforms involved in the two oxidative steps in the bioactivation of clopidogrel to its pharmacologically active metabolite. Clopidogrel 146-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 19812348-1 2010 The aim of the current study is to identify the human cytochrome P450 (P450) isoforms involved in the two oxidative steps in the bioactivation of clopidogrel to its pharmacologically active metabolite. Clopidogrel 146-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 19812348-2 2010 In the in vitro experiments using cDNA-expressed human P450 isoforms, clopidogrel was metabolized to 2-oxo-clopidogrel, the immediate precursor of its pharmacologically active metabolite. Clopidogrel 70-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 19812348-2 2010 In the in vitro experiments using cDNA-expressed human P450 isoforms, clopidogrel was metabolized to 2-oxo-clopidogrel, the immediate precursor of its pharmacologically active metabolite. 2-oxo-clopidogrel 101-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 19812348-7 2010 The contribution of CYP1A2, CYP2B6, and CYP2C19 to the formation of 2-oxo-clopidogrel was 35.8, 19.4, and 44.9%, respectively. 2-oxo-clopidogrel 68-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 19812351-9 2010 Constitutive expression of the tamoxifen-metabolizing enzymes CYP1B1, CYP2A6, CYP2B6, CYP2C8/9/19, CYP2D6, and SULT2A1 in glandular and surface epithelia was shown, but there was a large interindividual variation. Tamoxifen 31-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 20610889-2 2010 We investigated the relationship between CYP2B6 haplotype and incidence of TP-induced hepatotoxicity in 114 Japanese patients. Ticlopidine 75-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 19812066-8 2009 CONCLUSIONS: The CYP2B6 516G>T impact on nevirapine concentrations was less pronounced after intra-partum single-dose nevirapine than reported under steady-state conditions, perhaps due to lack of enzyme auto-induction at the time of dosing. Nevirapine 44-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 20001610-6 2010 The CYP2B6 G to T polymorphism at position 516 is shown to be associated with elevated plasma concentrations and an increase in neurotoxicity of EFV, while the wild-type genotype has been associated with sub-therapeutic concentrations of EFV, potentially leading to the development of viral resistance. efavirenz 145-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 20001610-6 2010 The CYP2B6 G to T polymorphism at position 516 is shown to be associated with elevated plasma concentrations and an increase in neurotoxicity of EFV, while the wild-type genotype has been associated with sub-therapeutic concentrations of EFV, potentially leading to the development of viral resistance. efavirenz 238-241 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 20017669-7 2010 In particular, for the CYP2B6 c.983T>C SNP, the difference in the alanine aminotransferase mean values were highly significant between TT and TC genotypes (p < 0.001). Technetium 145-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 19947890-5 2010 AREAS COVERED IN THIS REVIEW: In this communication, attempts have been made to bring together past as well as present information indicating that i) the P450 active site has two differently accessible allosterically interacting subsites geared for entirely different types of functionally relevant interactions; and ii) substrate binding to the specific protein residues (Site I) forming the reducible high-spin complex and product binding at L(6) (Site II) of the heme iron forming inhibited low-spin complex can regulate the functional state of the enzyme during catalysis. Heme 466-470 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 19947890-5 2010 AREAS COVERED IN THIS REVIEW: In this communication, attempts have been made to bring together past as well as present information indicating that i) the P450 active site has two differently accessible allosterically interacting subsites geared for entirely different types of functionally relevant interactions; and ii) substrate binding to the specific protein residues (Site I) forming the reducible high-spin complex and product binding at L(6) (Site II) of the heme iron forming inhibited low-spin complex can regulate the functional state of the enzyme during catalysis. Iron 471-475 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 19812066-8 2009 CONCLUSIONS: The CYP2B6 516G>T impact on nevirapine concentrations was less pronounced after intra-partum single-dose nevirapine than reported under steady-state conditions, perhaps due to lack of enzyme auto-induction at the time of dosing. Nevirapine 121-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 19708688-2 2009 The active oxidants in P450s have long been assumed to be iron(IV)-oxo porphyrin radical cations termed Compounds I, but P450 Compounds I have proven to be difficult to prepare. ferryl iron 58-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 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 2 subfamily B member 6 Homo sapiens 31-37 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 2 subfamily B member 6 Homo sapiens 130-136 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 2 subfamily B member 6 Homo sapiens 130-136 19608694-8 2009 The highest concentration of milnacipran (30 microM; >10 times plasma C(max)) produced 2.6- and 2.2-fold increases in CYP2B6 and CYP3A4/5 activity (making it 26 and 34% as effective as phenobarbital and rifampin, respectively). Milnacipran 29-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-127 20137387-0 2009 [Association between CYP2B6, CYP2D6, GSTP1 genetic polymorphisms and urinary styrene metabolites in professional workers]. Styrene 77-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 20137387-4 2009 RESULTS: The level of urine styrene metabolites level was influenced by genotypes of CYP2B6, CYP2D6 and GSTP1 [(280.28 +/- 100.60) mg/g Cr vs (183.48 +/- 127.52) mg/g Cr, (233.04 +/- 77.56) mg/g Cr vs (152.46 +/- 95.47) mg/g Cr, (32.88 +/- 7.14) mg/g Cr vs (24.47 +/- 5.59) mg/g Cr, P < 0.05)]. Styrene 28-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20137387-4 2009 RESULTS: The level of urine styrene metabolites level was influenced by genotypes of CYP2B6, CYP2D6 and GSTP1 [(280.28 +/- 100.60) mg/g Cr vs (183.48 +/- 127.52) mg/g Cr, (233.04 +/- 77.56) mg/g Cr vs (152.46 +/- 95.47) mg/g Cr, (32.88 +/- 7.14) mg/g Cr vs (24.47 +/- 5.59) mg/g Cr, P < 0.05)]. Chromium 136-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20137387-4 2009 RESULTS: The level of urine styrene metabolites level was influenced by genotypes of CYP2B6, CYP2D6 and GSTP1 [(280.28 +/- 100.60) mg/g Cr vs (183.48 +/- 127.52) mg/g Cr, (233.04 +/- 77.56) mg/g Cr vs (152.46 +/- 95.47) mg/g Cr, (32.88 +/- 7.14) mg/g Cr vs (24.47 +/- 5.59) mg/g Cr, P < 0.05)]. Chromium 167-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20137387-4 2009 RESULTS: The level of urine styrene metabolites level was influenced by genotypes of CYP2B6, CYP2D6 and GSTP1 [(280.28 +/- 100.60) mg/g Cr vs (183.48 +/- 127.52) mg/g Cr, (233.04 +/- 77.56) mg/g Cr vs (152.46 +/- 95.47) mg/g Cr, (32.88 +/- 7.14) mg/g Cr vs (24.47 +/- 5.59) mg/g Cr, P < 0.05)]. Chromium 167-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20137387-4 2009 RESULTS: The level of urine styrene metabolites level was influenced by genotypes of CYP2B6, CYP2D6 and GSTP1 [(280.28 +/- 100.60) mg/g Cr vs (183.48 +/- 127.52) mg/g Cr, (233.04 +/- 77.56) mg/g Cr vs (152.46 +/- 95.47) mg/g Cr, (32.88 +/- 7.14) mg/g Cr vs (24.47 +/- 5.59) mg/g Cr, P < 0.05)]. Chromium 167-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20137387-4 2009 RESULTS: The level of urine styrene metabolites level was influenced by genotypes of CYP2B6, CYP2D6 and GSTP1 [(280.28 +/- 100.60) mg/g Cr vs (183.48 +/- 127.52) mg/g Cr, (233.04 +/- 77.56) mg/g Cr vs (152.46 +/- 95.47) mg/g Cr, (32.88 +/- 7.14) mg/g Cr vs (24.47 +/- 5.59) mg/g Cr, P < 0.05)]. Chromium 167-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20137387-4 2009 RESULTS: The level of urine styrene metabolites level was influenced by genotypes of CYP2B6, CYP2D6 and GSTP1 [(280.28 +/- 100.60) mg/g Cr vs (183.48 +/- 127.52) mg/g Cr, (233.04 +/- 77.56) mg/g Cr vs (152.46 +/- 95.47) mg/g Cr, (32.88 +/- 7.14) mg/g Cr vs (24.47 +/- 5.59) mg/g Cr, P < 0.05)]. Chromium 167-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 20137387-5 2009 The metabolism of CYP2B6 G/G homozygotic genotype to styrene was more active than G/T heterozygotic genotype and T/T mutation genotype. Styrene 53-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 19693007-0 2009 Association of CYP2B6, CYP3A5, and CYP2C19 genetic polymorphisms with sibutramine pharmacokinetics in healthy Korean subjects. sibutramine 70-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 19693007-1 2009 We assessed the association of CYP2B6, CYP3A5, and CYP2C19 polymorphisms with sibutramine pharmacokinetics. sibutramine 78-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 19704027-8 2009 The oxidation of clopidogrel to its thiolactone metabolite correlated with variable activities of CYP1A2, CYP2B6, and CYP2C19. Clopidogrel 17-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 19704027-8 2009 The oxidation of clopidogrel to its thiolactone metabolite correlated with variable activities of CYP1A2, CYP2B6, and CYP2C19. Thiolactone 36-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 19198839-3 2009 RESULTS: Letrozole was a potent competitive inhibitor of CYP2A6 (K (i) 4.6 +/- 0.05 microM and 5.0 +/- 2.4 microM in HLMs and CYP2A6, respectively) and a weak inhibitor of CYP2C19 (K (i) 42.2 microM in HLMs and 33.3 microM in CYP2C19), while its metabolite showed moderate inhibition of CYP2C19 and CYP2B6. Letrozole 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 299-305 20137387-8 2009 CONCLUSION: Genotypes of CYP2B6, GSTP1 and CYP2D6 are related to susceptibility to the metabolism of styrene in human. Styrene 101-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 19708688-2 2009 The active oxidants in P450s have long been assumed to be iron(IV)-oxo porphyrin radical cations termed Compounds I, but P450 Compounds I have proven to be difficult to prepare. oxo porphyrin 67-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. Cyclophosphamide 87-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. Ifosfamide 105-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. Tamoxifen 117-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. Ketamine 128-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. artemisinin 138-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. Nevirapine 151-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. efavirenz 163-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. Bupropion 174-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19487249-0 2009 Inhibition of human CYP2B6-catalyzed bupropion hydroxylation by Ginkgo biloba extract: effect of terpene trilactones and flavonols. Bupropion 37-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. sibutramine 185-196 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19487249-0 2009 Inhibition of human CYP2B6-catalyzed bupropion hydroxylation by Ginkgo biloba extract: effect of terpene trilactones and flavonols. terpene trilactones 97-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 19487249-0 2009 Inhibition of human CYP2B6-catalyzed bupropion hydroxylation by Ginkgo biloba extract: effect of terpene trilactones and flavonols. Flavonols 121-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 19702527-4 2009 CYP2B6 can metabolise approximately 8% of clinically used drugs (n > 60), including cyclophosphamide, ifosfamide, tamoxifen, ketamine, artemisinin, nevirapine, efavirenz, bupropion, sibutramine, and propofol. Propofol 202-210 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19487249-5 2009 Therefore, we investigated the effect of G. biloba extract and some of its chemical constituents (terpene trilactones and flavonols) on the in vitro catalytic activity of CYP2B6 as assessed by the bupropion hydroxylation assay with recombinant enzyme and hepatic microsomes. terpene trilactones 98-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 19487249-5 2009 Therefore, we investigated the effect of G. biloba extract and some of its chemical constituents (terpene trilactones and flavonols) on the in vitro catalytic activity of CYP2B6 as assessed by the bupropion hydroxylation assay with recombinant enzyme and hepatic microsomes. Flavonols 122-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 19487249-5 2009 Therefore, we investigated the effect of G. biloba extract and some of its chemical constituents (terpene trilactones and flavonols) on the in vitro catalytic activity of CYP2B6 as assessed by the bupropion hydroxylation assay with recombinant enzyme and hepatic microsomes. Bupropion 197-206 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 19702527-7 2009 Typical substrates of CYP2B6 are non-planar molecules, neutral or weakly basic, highly lipophilic with one or two hydrogen-bond acceptors. Hydrogen 114-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 19487249-7 2009 Enzyme kinetic analysis indicated that G. biloba extract competitively inhibited hepatic microsomal CYP2B6-catalyzed bupropion hydroxylation (apparent K(i) was 162 +/- 14 microg/ml). Bupropion 117-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 19487249-10 2009 The inhibition of CYP2B6 by kaempferol was competitive (apparent K(i) was 10 +/- 1 microg/ml). kaempferol 28-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 19487249-11 2009 In summary, G. biloba extract and its flavonol aglycones are naturally occurring inhibitors of in vitro CYP2B6 catalytic activity and bupropion hydroxylation. 3-hydroxyflavone 38-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-110 19487251-7 2009 Incubation of lithocholic acid with a of human recombinant P450 enzymes revealed that all five metabolites were formed by recombinant CYP3A4. Lithocholic Acid 14-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-63 19487251-8 2009 Chemical inhibition studies with human liver microsomes and recombinant P450 enzymes confirmed that CYP3A4 was the predominant enzyme involved in hepatic microsomal biotransformation of lithocholic acid. Lithocholic Acid 186-202 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-76 19487251-9 2009 In summary, the results indicate that oxidation of the third carbon of the cholestane ring is the preferred position of oxidation by P450 enzymes for lithocholic acid biotransformation in humans and suggest that formation of lithocholic acid metabolites leads to enhanced hepatic detoxification and elimination. Carbon 61-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 19487251-9 2009 In summary, the results indicate that oxidation of the third carbon of the cholestane ring is the preferred position of oxidation by P450 enzymes for lithocholic acid biotransformation in humans and suggest that formation of lithocholic acid metabolites leads to enhanced hepatic detoxification and elimination. Cholestanes 75-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 19702527-18 2009 Smokers with the 1459C>T (R487C) variant of CYP2B6 may be more vulnerable to abstinence symptoms and relapse following treatment with bupropion as a smoking cessation agent. Bupropion 137-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 19487251-9 2009 In summary, the results indicate that oxidation of the third carbon of the cholestane ring is the preferred position of oxidation by P450 enzymes for lithocholic acid biotransformation in humans and suggest that formation of lithocholic acid metabolites leads to enhanced hepatic detoxification and elimination. Lithocholic Acid 150-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 19487251-9 2009 In summary, the results indicate that oxidation of the third carbon of the cholestane ring is the preferred position of oxidation by P450 enzymes for lithocholic acid biotransformation in humans and suggest that formation of lithocholic acid metabolites leads to enhanced hepatic detoxification and elimination. Lithocholic Acid 225-241 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 19393727-3 2009 To further clarify the role of human cytochrome P450 (P450 or CYP) and N-acetyltransferase (NAT) enzymes in the bioactivation of DNBeP to genotoxic metabolites, we determined the genotoxicity of DNBeP using a variety of umu tester strains expressing human P450 and NAT enzymes. 3,6-dinitrobenzopyrene 129-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-65 19520773-5 2009 Real-time polymerase chain reaction analysis of human hepatocyte cultures revealed that MD induces the mRNA expression of CYP2B6, CYP3A4, UGT1A1, and multidrug resistance 1 in a concentration-related manner, with the maximal induction of CYP2B6 challenging that of the induction by rifampicin. Rifampin 282-292 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 19520774-6 2009 Laromustine caused direct inhibition of CYP2B6 and CYP3A4/5 (the two enzymes involved in C-7 formation) as well as of CYP2C19. laromustine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 19520774-7 2009 K(i) values were 125 microM for CYP2B6, 297 muM for CYP3A4/5, and 349 microM for CYP2C19 and were greater than the average clinical plasma C(max) of laromustine (25 microM). laromustine 149-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 19520774-10 2009 The results of this study suggest the laromustine has 1) negligible victim potential with respect to metabolism by cytochrome P450 enzymes, 2) negligible enzyme-inducing potential, and 3) the potential in some cases to cause inhibition of CYP2B6, CYP3A4, and possibly CYP2C19 during and shortly after the duration of intravenous administration of this anticancer drug, but the clinical effects of such interactions are likely to be insignificant. laromustine 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 239-245 21217854-0 2009 Oxidation of the carcinogenic non-aminoazo dye 1-phenylazo-2-hydroxy-naphthalene (Sudan I) by cytochromes P450 and peroxidases: a comparative study. aminoazo dye 34-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-126 21217854-0 2009 Oxidation of the carcinogenic non-aminoazo dye 1-phenylazo-2-hydroxy-naphthalene (Sudan I) by cytochromes P450 and peroxidases: a comparative study. 1-phenylazo-2-naphthol 47-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-126 19084030-3 2009 Several reports have established that exposure to arsenite modifies P450 expression by decreasing or increasing mRNA and protein levels. arsenite 50-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 19393727-3 2009 To further clarify the role of human cytochrome P450 (P450 or CYP) and N-acetyltransferase (NAT) enzymes in the bioactivation of DNBeP to genotoxic metabolites, we determined the genotoxicity of DNBeP using a variety of umu tester strains expressing human P450 and NAT enzymes. 3,6-dinitrobenzopyrene 129-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 256-268 19393727-3 2009 To further clarify the role of human cytochrome P450 (P450 or CYP) and N-acetyltransferase (NAT) enzymes in the bioactivation of DNBeP to genotoxic metabolites, we determined the genotoxicity of DNBeP using a variety of umu tester strains expressing human P450 and NAT enzymes. 3,6-dinitrobenzopyrene 195-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-65 19454483-1 2009 17alpha-Ethinyl estradiol (EE) was systematically evaluated as a reversible and time-dependent inhibitor of 11 human drug-metabolizing cytochromes P450 (P450s) (CYP1A1, CYP1A2, CYP1B1, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2J2, CYP3A4, and CYP3A5) in vitro. Ethinyl Estradiol 0-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 185-191 19562679-0 2009 Artemisinin--a possible CYP2B6 probe substrate? artemisinin 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 19702548-1 2009 The present study demonstrated that in addition to CYP3A4 and CYP2D6, the metabolism of loratadine is also catalyzed by CYP1A1, CYP2C19, and to a lesser extent by CYP1A2, CYP2B6, CYP2C8, CYP2C9 and CYP3A5. Loratadine 88-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 19531981-1 2009 Cytochrome P450 2B6 (CYP2B6) is the main metabolizing pathway for efavirenz (EFV), the prescription of which is associated with neurologic side effects. efavirenz 66-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 19531981-1 2009 Cytochrome P450 2B6 (CYP2B6) is the main metabolizing pathway for efavirenz (EFV), the prescription of which is associated with neurologic side effects. efavirenz 66-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 19531981-1 2009 Cytochrome P450 2B6 (CYP2B6) is the main metabolizing pathway for efavirenz (EFV), the prescription of which is associated with neurologic side effects. efavirenz 77-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 19531981-1 2009 Cytochrome P450 2B6 (CYP2B6) is the main metabolizing pathway for efavirenz (EFV), the prescription of which is associated with neurologic side effects. efavirenz 77-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 19530977-2 2009 Since cytochrome P450 enzymes CYP3A4 and CYP2B6 play a major role in prasugrel"s active metabolite formation, the effect of potent CYP induction by rifampin on the pharmacokinetics of prasugrel and on the pharmacodynamic response to prasugrel was evaluated in healthy male subjects. Prasugrel Hydrochloride 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 19562679-3 2009 Correlations between the metabolic rate constant for artemisinin and the other CYP2B6 substrates were examined. artemisinin 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 19562679-8 2009 CONCLUSIONS: The rate of in vitro metabolism of artemisinin was correlated significantly to that of bupropion, propofol and efavirenz, suggesting artemisinin to be a potential alternative marker to assess CYP2B6 activity. artemisinin 48-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 205-211 19562679-9 2009 Further studies characterizing the metabolic fate of artemisinin are needed in order to evaluate its utility as an in vitro and in vivo CYP2B6 probe substrate, since CYP2B6 might not be the only CYP isoform involved in the depletion of artemisinin. artemisinin 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 19562679-9 2009 Further studies characterizing the metabolic fate of artemisinin are needed in order to evaluate its utility as an in vitro and in vivo CYP2B6 probe substrate, since CYP2B6 might not be the only CYP isoform involved in the depletion of artemisinin. artemisinin 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 19378397-4 2009 With cisplatin and oxaliplatin, a minor inhibition of CYP2C9 enzyme (75% of control at 400 miromol/l of these complexes) was seen; cisplatin also inhibited slightly the CYP2B6 activity (85% of control). Cisplatin 5-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 19409404-6 2009 Differentially expressed genes included the Phase I xenobiotic, fatty acid, sterol and steroid metabolism genes Cyp2b2 and CYP2B6, Cyp3a1 and CYP3A4, and Cyp4a22 and CYP4A11; Phase II conjugation enzyme genes Ugt1a1 and UGT1A1; and Phase III ABC transporter genes Abcb1 and ABCB1. Fatty Acids 64-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-129 19409404-6 2009 Differentially expressed genes included the Phase I xenobiotic, fatty acid, sterol and steroid metabolism genes Cyp2b2 and CYP2B6, Cyp3a1 and CYP3A4, and Cyp4a22 and CYP4A11; Phase II conjugation enzyme genes Ugt1a1 and UGT1A1; and Phase III ABC transporter genes Abcb1 and ABCB1. Sterols 76-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-129 19409404-6 2009 Differentially expressed genes included the Phase I xenobiotic, fatty acid, sterol and steroid metabolism genes Cyp2b2 and CYP2B6, Cyp3a1 and CYP3A4, and Cyp4a22 and CYP4A11; Phase II conjugation enzyme genes Ugt1a1 and UGT1A1; and Phase III ABC transporter genes Abcb1 and ABCB1. Steroids 87-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-129 19534586-4 2009 Truncated P450s 2A13, 2C9 (delta 3-20), 2C19 (delta 3-20), 2D6 (DB11) and 2E1 remained principally membrane-bound, but some P450 was found in the soluble fraction and could be partially extracted by alkaline and high salt treatments. Salts 217-221 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-14 19534586-6 2009 The MALLLAVFL modified forms of P450 2C9 YFP, P450 2C18 YFP and P450 2C19 YFP were found primarily at the periphery of the cells, whereas the truncated forms of P450 2C9 (delta 3-20) YFP and 2C19 (delta 3-20) YFP were observed at the periphery as well as inside the cells. malllavfl 4-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 19534586-8 2009 Rates of diclofenac 4 -hydroxylation by P450 2C9 and luciferin H-EGE metabolism by P450 2C19 were higher for the MALLLAVFL-modified forms compared with the (delta 3-20) truncated forms despite supplementation of truncated form incubations with additional reductase. Diclofenac 9-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-44 19378397-4 2009 With cisplatin and oxaliplatin, a minor inhibition of CYP2C9 enzyme (75% of control at 400 miromol/l of these complexes) was seen; cisplatin also inhibited slightly the CYP2B6 activity (85% of control). Cisplatin 131-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 19428328-10 2009 Moreover, DEHP dose-dependently induced CYP2B6 in human primary hepatocyte cultures. Diethylhexyl Phthalate 10-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 19135037-0 2009 In vitro assessment of P450 induction potential of novel chemopreventive agents SR13668, 9-cis-UAB30, and pentamethychromanol in primary cultures of human hepatocytes. SR 13668 80-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 23675126-4 2009 Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6) and the metabolic effects of EFV have been described previously. efavirenz 102-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 23675126-4 2009 Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6) and the metabolic effects of EFV have been described previously. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-63 23675126-4 2009 Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6) and the metabolic effects of EFV have been described previously. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 23675126-4 2009 Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6) and the metabolic effects of EFV have been described previously. efavirenz 11-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-63 23675126-4 2009 Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6) and the metabolic effects of EFV have been described previously. efavirenz 11-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 19135037-9 2009 Herein, we report that there is low potential for DDI with SR13668 and PMCol due to enzyme induction of CYP1A2, CYP2B6, and CYP3A4 expression at the concentrations examined. SR 13668 59-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-118 19135037-9 2009 Herein, we report that there is low potential for DDI with SR13668 and PMCol due to enzyme induction of CYP1A2, CYP2B6, and CYP3A4 expression at the concentrations examined. 2,2,5,7,8-pentamethyl-1-hydroxychroman 71-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-118 19135037-0 2009 In vitro assessment of P450 induction potential of novel chemopreventive agents SR13668, 9-cis-UAB30, and pentamethychromanol in primary cultures of human hepatocytes. UAB 30 89-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 19135037-0 2009 In vitro assessment of P450 induction potential of novel chemopreventive agents SR13668, 9-cis-UAB30, and pentamethychromanol in primary cultures of human hepatocytes. pentamethychromanol 106-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 19135037-6 2009 Therefore, a select panel of inducible P450 target genes (CYP1A2, CYP2B6, and CYP3A4) and their induction activity (measured by LC-MS/MS of respective marker substrate metabolites) were monitored in cultured hepatocytes following treatment with SR13668, 9-cis-UAB30, or PMCol to predict clinically significant drug-induced expression. SR 13668 245-252 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 19135037-6 2009 Therefore, a select panel of inducible P450 target genes (CYP1A2, CYP2B6, and CYP3A4) and their induction activity (measured by LC-MS/MS of respective marker substrate metabolites) were monitored in cultured hepatocytes following treatment with SR13668, 9-cis-UAB30, or PMCol to predict clinically significant drug-induced expression. UAB 30 254-265 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 19135037-6 2009 Therefore, a select panel of inducible P450 target genes (CYP1A2, CYP2B6, and CYP3A4) and their induction activity (measured by LC-MS/MS of respective marker substrate metabolites) were monitored in cultured hepatocytes following treatment with SR13668, 9-cis-UAB30, or PMCol to predict clinically significant drug-induced expression. 2,2,5,7,8-pentamethyl-1-hydroxychroman 270-275 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 19223781-1 2009 SETTING: Rifampicin may reduce plasma efavirenz concentrations by inducing the expression of the cytochrome P450 2B6, which metabolizes efavirenz. Rifampin 9-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-116 19228205-0 2009 Cytochrome P450 2B6 516G-->T is associated with plasma concentrations of nevirapine at both 200 mg twice daily and 400 mg once daily in an ethnically diverse population. Nevirapine 76-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 19228205-1 2009 OBJECTIVES: The aim of the study was to characterize the impact of the cytochrome P450 2B6 (CYP2B6), CYP3A4, CYP3A5 and ATP-binding cassette sub-family B member 1 (ABCB1) polymorphisms on nevirapine plasma concentrations. Nevirapine 188-198 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-90 19228205-1 2009 OBJECTIVES: The aim of the study was to characterize the impact of the cytochrome P450 2B6 (CYP2B6), CYP3A4, CYP3A5 and ATP-binding cassette sub-family B member 1 (ABCB1) polymorphisms on nevirapine plasma concentrations. Nevirapine 188-198 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 19228205-6 2009 In a multivariable analysis, CYP2B6 516G-->T and non-Caucasian ethnicity remained significant predictors of nevirapine C(trough) but CYP2B6 516G-->T homozygosity had the greatest effect (108% higher, 46% higher). nevirapine c 111-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 19228205-9 2009 The association between CYP2B6 516G-->T and higher plasma nevirapine exposure was maintained at both bid and qd dosing. Nevirapine 61-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 19171677-1 2009 Cytochrome P450 (P450) enzymes typically require the presence of at least cytochrome P450 reductase (CPR) and NADPH to carry out the metabolism of xenobiotics. NADP 110-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 19144770-0 2009 Mechanistic analysis of the inactivation of cytochrome P450 2B6 by phencyclidine: effects on substrate binding, electron transfer, and uncoupling. Phencyclidine 67-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-63 19371316-10 2009 Multiple linear regression analysis indicated that the CYP2B6 c.516G-->T polymorphism and CYP2A6 slow-metabolizing variants accounted for as much as 36 and 12% of the total variance in efavirenz concentrations, respectively. efavirenz 188-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 19158313-0 2009 Human pregnane X receptor activation and CYP3A4/CYP2B6 induction by 2,3-oxidosqualene:lanosterol cyclase inhibition. 2,3-oxidosqualene 68-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-54 19168709-0 2009 Metabolic activation of mifepristone [RU486; 17beta-hydroxy-11beta-(4-dimethylaminophenyl)-17alpha-(1-propynyl)-estra-4,9-dien-3-one] by mammalian cytochromes P450 and the mechanism-based inactivation of human CYP2B6. Mifepristone 24-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 210-216 19601870-4 2009 The analysis of P450-catalyzed reaction rates is elaborated to encompass a treatment of metabolic clearance, and satisfactory correlations are obtained with literature values for both intrinsic clearance and whole body clearance in terms of compound lipophilicity derived from log P data, where P is the octanol/water partition coefficient. Octanols 304-311 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 19601870-4 2009 The analysis of P450-catalyzed reaction rates is elaborated to encompass a treatment of metabolic clearance, and satisfactory correlations are obtained with literature values for both intrinsic clearance and whole body clearance in terms of compound lipophilicity derived from log P data, where P is the octanol/water partition coefficient. Water 312-317 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 19168709-0 2009 Metabolic activation of mifepristone [RU486; 17beta-hydroxy-11beta-(4-dimethylaminophenyl)-17alpha-(1-propynyl)-estra-4,9-dien-3-one] by mammalian cytochromes P450 and the mechanism-based inactivation of human CYP2B6. 17beta-hydroxy-11beta-(4-dimethylaminophenyl)-17alpha-(1-propynyl)-estra-4,9-dien-3-one 45-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 210-216 19168709-1 2009 Mifepristone [RU486; 17beta-hydroxy-11beta-(4-dimethylaminophenyl)-17alpha-(1-propynyl)-estra-4,9-dien-3-one] inactivates CYP2B6 in the reconstituted system in a mechanism-based manner. Mifepristone 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 19168709-1 2009 Mifepristone [RU486; 17beta-hydroxy-11beta-(4-dimethylaminophenyl)-17alpha-(1-propynyl)-estra-4,9-dien-3-one] inactivates CYP2B6 in the reconstituted system in a mechanism-based manner. 17beta-hydroxy-11beta-(4-dimethylaminophenyl)-17alpha-(1-propynyl)-estra-4,9-dien-3-one 21-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 19168709-2 2009 The loss of 7-ethoxy-4-(trifluoromethyl)-coumarin deethylation activity of CYP2B6 is concentration- and time-dependent. 7-ethoxy-4-(trifluoromethyl) 12-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 19168709-2 2009 The loss of 7-ethoxy-4-(trifluoromethyl)-coumarin deethylation activity of CYP2B6 is concentration- and time-dependent. coumarin 41-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 19168709-5 2009 Incubation of CYP2B6 with 20 microM RU486 for 15 min resulted in 61% loss of catalytic activity, 60% loss of the reduced cytochrome P450 (P450)-CO complex, and a 40% loss of native heme. Mifepristone 36-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 19168709-5 2009 Incubation of CYP2B6 with 20 microM RU486 for 15 min resulted in 61% loss of catalytic activity, 60% loss of the reduced cytochrome P450 (P450)-CO complex, and a 40% loss of native heme. Heme 181-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 19168709-7 2009 SDS-polyacrylamide gel electrophoresis and high-pressure liquid chromatography analysis showed that [(3)H]RU486 was irreversibly bound to CYP2B6 apoprotein. sds-polyacrylamide 0-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 19168709-7 2009 SDS-polyacrylamide gel electrophoresis and high-pressure liquid chromatography analysis showed that [(3)H]RU486 was irreversibly bound to CYP2B6 apoprotein. Mifepristone 106-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 19168709-13 2009 It seems that the potential for drug-drug interactions of RU486 may not be limited only to CYP3A4 and should also be evaluated for drugs metabolized primarily by CYP2B6, such as bupropion and efavirenz. Mifepristone 58-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. ginkgolide A 147-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. ginkgolide B 155-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. BDBM50323769 124-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. ginkgolide B 169-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. bilobalide 185-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. boeravinone B 197-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. Flavonoids 208-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. Quercetin 220-229 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. kaempferol 231-241 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. tamarixetin 246-257 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 19238117-4 2009 We hypothesize that genetic variability in this gene may contribute to the particularly high, unexplained variability in EFV exposure in individuals with limited CYP2B6 function. efavirenz 121-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 19239339-1 2009 BACKGROUND: Polymorphisms in CYP2B6 affect the steady-state plasma concentrations of nevirapine and efavirenz. Nevirapine 85-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 19239339-1 2009 BACKGROUND: Polymorphisms in CYP2B6 affect the steady-state plasma concentrations of nevirapine and efavirenz. efavirenz 100-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 19239339-10 2009 The composite CYP2B6 516/983 genotype was significantly associated with plasma drug exposure and clearance for efavirenz but not nevirapine. efavirenz 111-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 19239339-11 2009 Exploratory analyses suggested possible associations between additional CYP2B6 polymorphisms and the pharmacokinetics of nevirapine and efavirenz. Nevirapine 121-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 19074527-5 2009 Likewise, K262R showed 2-, 4-, and >20-fold higher K(s) values than CYP2B6dH with clopidogrel, sertraline, and itraconazole, respectively. k262r 10-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 19442086-6 2009 CYP2B6, CYP3A4 and CYP2C9 play a relevant role in the metabolism of ketamine. Ketamine 68-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19442086-8 2009 Propofol is metabolized mainly by glucuronidation by uridine diphosphate-glucuronosyltransferases (UGTs) and by hydroxylation by CYP2B6 and CYP2C enzymes. Propofol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-135 19047469-0 2009 Mechanism-based inhibition of human cytochrome P450 2B6 by ticlopidine, clopidogrel, and the thiolactone metabolite of prasugrel. Ticlopidine 59-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-55 19047469-0 2009 Mechanism-based inhibition of human cytochrome P450 2B6 by ticlopidine, clopidogrel, and the thiolactone metabolite of prasugrel. Clopidogrel 72-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-55 19074527-5 2009 Likewise, K262R showed 2-, 4-, and >20-fold higher K(s) values than CYP2B6dH with clopidogrel, sertraline, and itraconazole, respectively. Clopidogrel 85-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 19047469-0 2009 Mechanism-based inhibition of human cytochrome P450 2B6 by ticlopidine, clopidogrel, and the thiolactone metabolite of prasugrel. Thiolactone 93-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-55 19047469-0 2009 Mechanism-based inhibition of human cytochrome P450 2B6 by ticlopidine, clopidogrel, and the thiolactone metabolite of prasugrel. Prasugrel Hydrochloride 119-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-55 19074527-5 2009 Likewise, K262R showed 2-, 4-, and >20-fold higher K(s) values than CYP2B6dH with clopidogrel, sertraline, and itraconazole, respectively. Sertraline 98-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 19047469-1 2009 Mechanism-based inhibition of CYP2B6 in human liver microsomes by thienopyridine antiplatelet agents ticlopidine and clopidogrel and the thiolactone metabolites of those two agents plus that of prasugrel were investigated by determining the time- and concentration-dependent inhibition of the activity of bupropion hydroxylase as the typical CYP2B6 activity. thienopyridine 66-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 19022226-5 2009 The chemical CYP2B6 inhibitor 4-(4-chlorobenzyl)pyridine (CBP) reduced the metabolite formation in pooled HLM by 63% at 1 microM PCEEA. 4-(4-Chlorobenzyl)pyridine 30-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 19047469-1 2009 Mechanism-based inhibition of CYP2B6 in human liver microsomes by thienopyridine antiplatelet agents ticlopidine and clopidogrel and the thiolactone metabolites of those two agents plus that of prasugrel were investigated by determining the time- and concentration-dependent inhibition of the activity of bupropion hydroxylase as the typical CYP2B6 activity. Ticlopidine 101-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 19047469-1 2009 Mechanism-based inhibition of CYP2B6 in human liver microsomes by thienopyridine antiplatelet agents ticlopidine and clopidogrel and the thiolactone metabolites of those two agents plus that of prasugrel were investigated by determining the time- and concentration-dependent inhibition of the activity of bupropion hydroxylase as the typical CYP2B6 activity. Clopidogrel 117-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 19047469-2 2009 By comparing the ratios of k(inact) (maximal inactivation rate constant)/K(I) (the inactivator concentration producing a half-maximal rate of inactivation), it was found that the thiolactone metabolite of prasugrel is 10- and 22-fold less potent, respectively, in the mechanism-based inhibition of CYP2B6 than ticlopidine and clopidogrel. Thiolactone 179-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 298-304 19047469-2 2009 By comparing the ratios of k(inact) (maximal inactivation rate constant)/K(I) (the inactivator concentration producing a half-maximal rate of inactivation), it was found that the thiolactone metabolite of prasugrel is 10- and 22-fold less potent, respectively, in the mechanism-based inhibition of CYP2B6 than ticlopidine and clopidogrel. Prasugrel Hydrochloride 205-214 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 298-304 19047469-4 2009 In conclusion, ticlopidine, its thiolactone metabolite, and clopidogrel were more potent mechanism-based inhibitors of CYP2B6 than the thiolactone metabolite of prasugrel. Ticlopidine 15-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 19047469-4 2009 In conclusion, ticlopidine, its thiolactone metabolite, and clopidogrel were more potent mechanism-based inhibitors of CYP2B6 than the thiolactone metabolite of prasugrel. Thiolactone 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 19047469-4 2009 In conclusion, ticlopidine, its thiolactone metabolite, and clopidogrel were more potent mechanism-based inhibitors of CYP2B6 than the thiolactone metabolite of prasugrel. Clopidogrel 60-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 19047469-4 2009 In conclusion, ticlopidine, its thiolactone metabolite, and clopidogrel were more potent mechanism-based inhibitors of CYP2B6 than the thiolactone metabolite of prasugrel. Thiolactone 135-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 19047469-4 2009 In conclusion, ticlopidine, its thiolactone metabolite, and clopidogrel were more potent mechanism-based inhibitors of CYP2B6 than the thiolactone metabolite of prasugrel. Prasugrel Hydrochloride 161-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 19005482-7 2009 Among other clinical and genetic factors, polymorphisms of CYP2B6 genes that interfere with cyclophosphamide metabolism were associated with OM (recipient CYP2B6(*)4; P=0.0067), HC (recipient CYP2B6(*)2; P=0.03) and VOD (donor CYP2B6(*)6; P=0.03). Cyclophosphamide 92-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 19005482-7 2009 Among other clinical and genetic factors, polymorphisms of CYP2B6 genes that interfere with cyclophosphamide metabolism were associated with OM (recipient CYP2B6(*)4; P=0.0067), HC (recipient CYP2B6(*)2; P=0.03) and VOD (donor CYP2B6(*)6; P=0.03). Cyclophosphamide 92-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-161 19005482-7 2009 Among other clinical and genetic factors, polymorphisms of CYP2B6 genes that interfere with cyclophosphamide metabolism were associated with OM (recipient CYP2B6(*)4; P=0.0067), HC (recipient CYP2B6(*)2; P=0.03) and VOD (donor CYP2B6(*)6; P=0.03). Cyclophosphamide 92-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-161 19005482-7 2009 Among other clinical and genetic factors, polymorphisms of CYP2B6 genes that interfere with cyclophosphamide metabolism were associated with OM (recipient CYP2B6(*)4; P=0.0067), HC (recipient CYP2B6(*)2; P=0.03) and VOD (donor CYP2B6(*)6; P=0.03). Cyclophosphamide 92-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-161 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 2 subfamily B member 6 Homo sapiens 115-128 19029318-5 2009 In pilot experiments, voriconazole showed inhibition of CYP2B6, CYP2C9, CYP2C19, and CYP3A (half-maximal [50%] inhibitory concentrations, <6 microM); its effect on CYP1A2, CYP2A6, CYP2C8, and CYP2D6 was marginal (<25% inhibition at 100 microM voriconazole). Voriconazole 22-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 19029318-6 2009 Further detailed experiments with HLMs showed that voriconazole is a potent competitive inhibitor of CYP2B6 (K(i) < 0.5), CYP2C9 (K(i) = 2.79 microM), and CYP2C19 (K(i) = 5.1 microM). Voriconazole 51-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 19095658-11 2009 Together, these findings indicate that phosphorylation of CYP3A4 Ser(478), Thr(264), and Ser(420) residues by cytosolic kinases is important both for its ubiquitination and proteasomal degradation and suggest a direct link between P450 phosphorylation, ubiquitination, and degradation. Serine 65-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 231-235 19095658-11 2009 Together, these findings indicate that phosphorylation of CYP3A4 Ser(478), Thr(264), and Ser(420) residues by cytosolic kinases is important both for its ubiquitination and proteasomal degradation and suggest a direct link between P450 phosphorylation, ubiquitination, and degradation. Threonine 75-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 231-235 19095658-11 2009 Together, these findings indicate that phosphorylation of CYP3A4 Ser(478), Thr(264), and Ser(420) residues by cytosolic kinases is important both for its ubiquitination and proteasomal degradation and suggest a direct link between P450 phosphorylation, ubiquitination, and degradation. Serine 89-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 231-235 18976853-0 2009 Bioactivation of the phytoestrogen diosmetin by CYP1 cytochromes P450. diosmetin 35-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 18976853-3 2009 In this study we investigated the anticancer activity of the flavonoid diosmetin, as a result of cytochrome P450 CYP1 metabolism. Flavonoids 61-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 18976853-3 2009 In this study we investigated the anticancer activity of the flavonoid diosmetin, as a result of cytochrome P450 CYP1 metabolism. diosmetin 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 19022226-5 2009 The chemical CYP2B6 inhibitor 4-(4-chlorobenzyl)pyridine (CBP) reduced the metabolite formation in pooled HLM by 63% at 1 microM PCEEA. 4,4'-Bis(N-carbazolyl)-1,1'-biphenyl 58-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 18971317-9 2009 Rosuvastatin was found to increase human constitutive androstane receptor (hCAR)-mediated transcription of CYP3A4, CYP2C9, and CYP2B6 genes, predicting the consequent potential for drug interactions with several coadministered drugs. Rosuvastatin Calcium 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 18971317-10 2009 Activation of hCAR and hPXR by atorvastatin and the subsequent induction of not only CYP2B6 and CYP3A4 but also of CYP2C9 present an additional target by which atorvastatin, a widely used cholesterol-lowering drug, can modify the kinetics of numerous drugs. Atorvastatin 160-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 18971317-10 2009 Activation of hCAR and hPXR by atorvastatin and the subsequent induction of not only CYP2B6 and CYP3A4 but also of CYP2C9 present an additional target by which atorvastatin, a widely used cholesterol-lowering drug, can modify the kinetics of numerous drugs. Cholesterol 188-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 18983865-6 2009 The strong and overlapping inductive role of phenobarbital strengthens the participation of CYP2B6 and CYP3A in diazepam N-demethylation and CYP3A in temazepam formation. Phenobarbital 45-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 19017849-8 2009 Metabolite determinations of the dimethyl-ifosfamide analogs were performed using liquid chromatography and tandem mass spectrometry after in vitro biotransformation by drug-induced rat liver microsomes and human microsomes expressing the main CYP3A4 and minor CYP2B6 enzymes. dimethyl-ifosfamide 33-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 261-267 19135815-0 2009 Use of fission yeast heterologously expressing human cytochrome P450 2B6 in biotechnological synthesis of the designer drug metabolite N-(1-phenylcyclohexyl)-2-hydroxyethanamine. N-(1-phenylcyclohexyl)-2-hydroxyethanamine 135-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-72 19029318-8 2009 Prediction of the in vivo interaction of voriconazole from these in vitro data suggests that voriconazole would substantially increase the exposure of drugs metabolized by CYP2B6, CYP2C9, CYP2C19, and CYP3A. Voriconazole 41-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-178 19029318-8 2009 Prediction of the in vivo interaction of voriconazole from these in vitro data suggests that voriconazole would substantially increase the exposure of drugs metabolized by CYP2B6, CYP2C9, CYP2C19, and CYP3A. Voriconazole 93-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-178 18837430-4 2009 Because of its metabolism by hepatic p450 enzymes, voriconazole may inhibit the clearance of many medications, including vincristine. Voriconazole 51-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-41 18837430-4 2009 Because of its metabolism by hepatic p450 enzymes, voriconazole may inhibit the clearance of many medications, including vincristine. Vincristine 121-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-41 18983865-6 2009 The strong and overlapping inductive role of phenobarbital strengthens the participation of CYP2B6 and CYP3A in diazepam N-demethylation and CYP3A in temazepam formation. Diazepam 112-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 19076156-0 2009 Polymorphisms of drug-metabolizing enzymes (GST, CYP2B6 and CYP3A) affect the pharmacokinetics of thiotepa and tepa. Thiotepa 98-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 19089010-1 2009 The mechanism of N-dealkylation of N-cyclopropyl-N-methylaniline () catalyzed by cytochrome P450 (P450) was investigated using density functional theory. Nitrogen 17-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-103 19089010-1 2009 The mechanism of N-dealkylation of N-cyclopropyl-N-methylaniline () catalyzed by cytochrome P450 (P450) was investigated using density functional theory. n-cyclopropyl-n-methylaniline 35-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-103 19704172-1 2009 BACKGROUND: Rifampicin induces expression of the cytochrome P450 isoenzyme 2B6 (CYP2B6), which metabolizes efavirenz. Rifampin 12-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-78 19704172-1 2009 BACKGROUND: Rifampicin induces expression of the cytochrome P450 isoenzyme 2B6 (CYP2B6), which metabolizes efavirenz. Rifampin 12-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 22190985-5 2009 The pronounced inhibitory impact of voriconazole on methadone metabolism via the cytochrome P450 (CYP)2B6 isoenzyme was identified as a probable cause of the arrhythmia. Voriconazole 36-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-105 22190985-5 2009 The pronounced inhibitory impact of voriconazole on methadone metabolism via the cytochrome P450 (CYP)2B6 isoenzyme was identified as a probable cause of the arrhythmia. Methadone 52-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-105 19076156-0 2009 Polymorphisms of drug-metabolizing enzymes (GST, CYP2B6 and CYP3A) affect the pharmacokinetics of thiotepa and tepa. Triethylenephosphoramide 102-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 19076156-5 2009 The purpose of this study was to evaluate effects of known allelic variants in CYP2B6, CYP3A4, CYP3A5, GSTA1 and GSTP1 genes on pharmacokinetics of thiotepa and tepa. Thiotepa 148-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 19076156-5 2009 The purpose of this study was to evaluate effects of known allelic variants in CYP2B6, CYP3A4, CYP3A5, GSTA1 and GSTP1 genes on pharmacokinetics of thiotepa and tepa. Triethylenephosphoramide 152-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 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 2 subfamily B member 6 Homo sapiens 21-27 19185039-0 2009 Mechanistic insight into formation of oxo-iron(IV) porphyrin pi-cation radicals from enzyme mimics of cytochrome P450 in organic solvents. oxo-iron 38-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-117 19185039-0 2009 Mechanistic insight into formation of oxo-iron(IV) porphyrin pi-cation radicals from enzyme mimics of cytochrome P450 in organic solvents. porphyrin pi cation radical 51-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-117 19185039-1 2009 Two new models for cytochrome P450 in which the thiolate axial ligand is replaced by a RSO(3)(-) group, form oxo-iron(IV) porphyrin pi-cation radicals as sole oxidation products in "peroxo shunt" reactions independent of the nature of the employed solvent (polar or non-polar) and electronic nature of the porphyrin rings. thiolate 48-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 19185039-1 2009 Two new models for cytochrome P450 in which the thiolate axial ligand is replaced by a RSO(3)(-) group, form oxo-iron(IV) porphyrin pi-cation radicals as sole oxidation products in "peroxo shunt" reactions independent of the nature of the employed solvent (polar or non-polar) and electronic nature of the porphyrin rings. (R)-Styrene oxide 87-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 19185039-1 2009 Two new models for cytochrome P450 in which the thiolate axial ligand is replaced by a RSO(3)(-) group, form oxo-iron(IV) porphyrin pi-cation radicals as sole oxidation products in "peroxo shunt" reactions independent of the nature of the employed solvent (polar or non-polar) and electronic nature of the porphyrin rings. oxo-iron 109-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 19185039-1 2009 Two new models for cytochrome P450 in which the thiolate axial ligand is replaced by a RSO(3)(-) group, form oxo-iron(IV) porphyrin pi-cation radicals as sole oxidation products in "peroxo shunt" reactions independent of the nature of the employed solvent (polar or non-polar) and electronic nature of the porphyrin rings. Porphyrins 122-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 19185039-3 2009 This article reports the results of mechanistic studies involving the measurements of the rate of oxo-iron(IV) porphyrin pi-cation radical formation from the enzyme mimics of P450 for different oxidant concentration, temperature and pressure in selected organic solvents. oxo-iron(iv) porphyrin 98-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 175-179 19474465-2 2009 Single nucleotide polymorphisms of the hepatic cytochrome P450 isoenzyme 2B6 (CYP2B6) gene have been associated with high interindividual variations in EFV plasma concentrations. efavirenz 152-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-76 19474465-2 2009 Single nucleotide polymorphisms of the hepatic cytochrome P450 isoenzyme 2B6 (CYP2B6) gene have been associated with high interindividual variations in EFV plasma concentrations. efavirenz 152-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 19474465-3 2009 Our objective was to determine the adequacy of EFV dosing and explore the influence of CYP2B6-516G>T polymorphisms on EFV plasma concentrations in Thai HIV-infected children. efavirenz 121-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 19474465-15 2009 CYP2B6-516G>T polymorphisms significantly affect the drug metabolism of EFV in children. efavirenz 75-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 19053182-5 2009 In the current study, we demonstrate that M1 is a stable metabolite that is highly susceptible to facile oxidation by cytochrome P450 enzymes (P450s) to form a reactive diiminoquinone intermediate (M2). diiminoquinone 169-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 19053182-8 2009 The formation of diiminoquinone M2 was P450 mediated with 2C19 and 1A2 as the two principal P450 enzymes catalyzing M1 oxidation. diiminoquinone 17-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 19053182-8 2009 The formation of diiminoquinone M2 was P450 mediated with 2C19 and 1A2 as the two principal P450 enzymes catalyzing M1 oxidation. diiminoquinone 17-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 19053182-10 2009 P450 2C19 exclusively mediated further metabolism of M1 to the amino hydroxynimesulide M3 and its diiminoquinone M4. amino hydroxynimesulide 63-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 19053182-10 2009 P450 2C19 exclusively mediated further metabolism of M1 to the amino hydroxynimesulide M3 and its diiminoquinone M4. diiminoquinone m4 98-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 19745566-4 2009 High P450 2D-dependent bufuralol 1"-hydroxylation and FMO-dependent benzydamine N-oxygenation activity was observed in liver microsomes from Microminipigs. bufuralol 23-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 19745566-6 2009 However, occasional differences might give undetected low P450 2A-dependent coumarin 7-hydroxylation in Microminipigs at 8-months-old, in contrast to liver microsomes from one 10-days-old Microminipis and commercially available pooled minipigs which had low but detectable coumarin 7-hydroxylation activity. coumarin 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 18989234-0 2008 Efavirenz induces CYP2B6-mediated hydroxylation of bupropion in healthy subjects. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 19209593-4 2009 Enantioselectivity was also observed for PCBs 45 (EF = 0.437) and 132 (EF = 0.537) incubated at that concentration with human CYP 2B6. Polychlorinated Biphenyls 41-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-133 18992768-4 2009 PGRMC1 and its homologues regulate cholesterol synthesis by activating the P450 protein Cyp51/lanosterol demethylase, and the cholesterol synthetic pathway is an important target in cardiovascular disease and in treating infections. Cholesterol 35-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 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 2 subfamily B member 6 Homo sapiens 198-204 18563875-0 2008 How do azoles inhibit cytochrome P450 enzymes? Azoles 7-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 18563875-2 2008 To examine how azole inhibitors interact with the heme active site of the cytochrome P450 enzymes, we have performed a series of density functional theory studies on azole binding. Azoles 15-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 18563875-2 2008 To examine how azole inhibitors interact with the heme active site of the cytochrome P450 enzymes, we have performed a series of density functional theory studies on azole binding. Heme 50-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 18563875-2 2008 To examine how azole inhibitors interact with the heme active site of the cytochrome P450 enzymes, we have performed a series of density functional theory studies on azole binding. Azoles 166-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 18563875-3 2008 These are the first density functional studies on azole interactions with a heme center and give fundamental insight into how azoles inhibit the catalytic function of P450 enzymes. Azoles 50-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-171 18563875-3 2008 These are the first density functional studies on azole interactions with a heme center and give fundamental insight into how azoles inhibit the catalytic function of P450 enzymes. Heme 76-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-171 18563875-3 2008 These are the first density functional studies on azole interactions with a heme center and give fundamental insight into how azoles inhibit the catalytic function of P450 enzymes. Azoles 126-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-171 18563875-6 2008 The calculations show that azole binding is a stepwise mechanism whereby first the water molecule from the resting state of P450 is released from the sixth binding site of the heme to create a pentacoordinated active site followed by coordination of the azole nitrogen to the heme iron. Azoles 27-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 18563875-6 2008 The calculations show that azole binding is a stepwise mechanism whereby first the water molecule from the resting state of P450 is released from the sixth binding site of the heme to create a pentacoordinated active site followed by coordination of the azole nitrogen to the heme iron. Water 83-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 18563875-6 2008 The calculations show that azole binding is a stepwise mechanism whereby first the water molecule from the resting state of P450 is released from the sixth binding site of the heme to create a pentacoordinated active site followed by coordination of the azole nitrogen to the heme iron. Heme 176-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 18563875-6 2008 The calculations show that azole binding is a stepwise mechanism whereby first the water molecule from the resting state of P450 is released from the sixth binding site of the heme to create a pentacoordinated active site followed by coordination of the azole nitrogen to the heme iron. Azoles 254-259 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 18563875-6 2008 The calculations show that azole binding is a stepwise mechanism whereby first the water molecule from the resting state of P450 is released from the sixth binding site of the heme to create a pentacoordinated active site followed by coordination of the azole nitrogen to the heme iron. Nitrogen 260-268 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 18563875-6 2008 The calculations show that azole binding is a stepwise mechanism whereby first the water molecule from the resting state of P450 is released from the sixth binding site of the heme to create a pentacoordinated active site followed by coordination of the azole nitrogen to the heme iron. Heme 276-280 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 18563875-6 2008 The calculations show that azole binding is a stepwise mechanism whereby first the water molecule from the resting state of P450 is released from the sixth binding site of the heme to create a pentacoordinated active site followed by coordination of the azole nitrogen to the heme iron. Iron 281-285 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 18563875-9 2008 Thus, we show that release of a water molecule from the resting state of P450 enzymes to create a pentacoordinated heme will lead to a doublet to quartet spin state crossing at an Fe-OH(2) distance of approximately 3.0 A, while the azole substitution process takes place at shorter distances. Water 32-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 18563875-9 2008 Thus, we show that release of a water molecule from the resting state of P450 enzymes to create a pentacoordinated heme will lead to a doublet to quartet spin state crossing at an Fe-OH(2) distance of approximately 3.0 A, while the azole substitution process takes place at shorter distances. Heme 115-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 18563875-9 2008 Thus, we show that release of a water molecule from the resting state of P450 enzymes to create a pentacoordinated heme will lead to a doublet to quartet spin state crossing at an Fe-OH(2) distance of approximately 3.0 A, while the azole substitution process takes place at shorter distances. fe-oh(2) 180-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 18563875-9 2008 Thus, we show that release of a water molecule from the resting state of P450 enzymes to create a pentacoordinated heme will lead to a doublet to quartet spin state crossing at an Fe-OH(2) distance of approximately 3.0 A, while the azole substitution process takes place at shorter distances. Azoles 232-237 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 18563875-12 2008 Studies with an extra hydrogen-bonded ethanol molecule in the model, mimicking the active site of the CYP121 P450, show that the resting state and azole binding structures are close in energy, which may lead to chemical equilibrium between the two structures, as indeed observed with recent protein structural studies that have demonstrated two distinct azole binding mechanisms to P450 heme. Hydrogen 22-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 18563875-12 2008 Studies with an extra hydrogen-bonded ethanol molecule in the model, mimicking the active site of the CYP121 P450, show that the resting state and azole binding structures are close in energy, which may lead to chemical equilibrium between the two structures, as indeed observed with recent protein structural studies that have demonstrated two distinct azole binding mechanisms to P450 heme. Hydrogen 22-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 382-386 18563875-12 2008 Studies with an extra hydrogen-bonded ethanol molecule in the model, mimicking the active site of the CYP121 P450, show that the resting state and azole binding structures are close in energy, which may lead to chemical equilibrium between the two structures, as indeed observed with recent protein structural studies that have demonstrated two distinct azole binding mechanisms to P450 heme. Ethanol 38-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 18563875-12 2008 Studies with an extra hydrogen-bonded ethanol molecule in the model, mimicking the active site of the CYP121 P450, show that the resting state and azole binding structures are close in energy, which may lead to chemical equilibrium between the two structures, as indeed observed with recent protein structural studies that have demonstrated two distinct azole binding mechanisms to P450 heme. Ethanol 38-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 382-386 18563875-12 2008 Studies with an extra hydrogen-bonded ethanol molecule in the model, mimicking the active site of the CYP121 P450, show that the resting state and azole binding structures are close in energy, which may lead to chemical equilibrium between the two structures, as indeed observed with recent protein structural studies that have demonstrated two distinct azole binding mechanisms to P450 heme. Azoles 147-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 18563875-12 2008 Studies with an extra hydrogen-bonded ethanol molecule in the model, mimicking the active site of the CYP121 P450, show that the resting state and azole binding structures are close in energy, which may lead to chemical equilibrium between the two structures, as indeed observed with recent protein structural studies that have demonstrated two distinct azole binding mechanisms to P450 heme. Azoles 147-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 382-386 18563875-12 2008 Studies with an extra hydrogen-bonded ethanol molecule in the model, mimicking the active site of the CYP121 P450, show that the resting state and azole binding structures are close in energy, which may lead to chemical equilibrium between the two structures, as indeed observed with recent protein structural studies that have demonstrated two distinct azole binding mechanisms to P450 heme. Azoles 354-359 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 18563875-12 2008 Studies with an extra hydrogen-bonded ethanol molecule in the model, mimicking the active site of the CYP121 P450, show that the resting state and azole binding structures are close in energy, which may lead to chemical equilibrium between the two structures, as indeed observed with recent protein structural studies that have demonstrated two distinct azole binding mechanisms to P450 heme. Heme 387-391 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 18989234-0 2008 Efavirenz induces CYP2B6-mediated hydroxylation of bupropion in healthy subjects. Bupropion 51-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 18989234-1 2008 OBJECTIVE: To characterize the effect of efavirenz on bupropion hydroxylation as a marker of cytochrome P450 (CYP) 2B6 activity in healthy subjects. Bupropion 54-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-118 18989234-9 2008 CONCLUSIONS: Our results confirm that efavirenz induces CYP2B6 enzyme activity in vivo, as demonstrated by an increase in bupropion hydroxylation after 2 weeks of efavirenz administration. efavirenz 38-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 18989234-9 2008 CONCLUSIONS: Our results confirm that efavirenz induces CYP2B6 enzyme activity in vivo, as demonstrated by an increase in bupropion hydroxylation after 2 weeks of efavirenz administration. Bupropion 122-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 18989234-9 2008 CONCLUSIONS: Our results confirm that efavirenz induces CYP2B6 enzyme activity in vivo, as demonstrated by an increase in bupropion hydroxylation after 2 weeks of efavirenz administration. efavirenz 163-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 18799803-3 2008 SKF525A and its metabolite and primary amine analog all inhibited CYP2B6-, CYP2C9-, CYP2C19-, CYP2D6-, and CYP3A-selective reactions to varying degrees but had little effect on CYP1A2, CYP2A6, and CYP2E1 reactions. Proadifen 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 18787056-8 2008 At a higher IBU enantiomer concentration (500 microM), additional P450s catalyzed 2-hydroxylation (CYP3A4, CYP2C8, CYP2C19, CYP2D6, CYP2E1, and CYP2B6) and 3-hydroxylation (CYP2C19). Ibuprofen 12-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-150 19356104-4 2008 Therefore, the methadone-maintenance-treatment outcome should be evaluated when patients are treated with drugs which are supposed to induce CYP3A4 and CYP2B6 isoforms. Methadone 15-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-158 18799803-3 2008 SKF525A and its metabolite and primary amine analog all inhibited CYP2B6-, CYP2C9-, CYP2C19-, CYP2D6-, and CYP3A-selective reactions to varying degrees but had little effect on CYP1A2, CYP2A6, and CYP2E1 reactions. Amines 39-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 18687804-7 2008 Spectral analysis of the interaction with P450s revealed that (-)-phenylahistin led to a hydrophobic type I signature, whereas the (+)-isomer yielded a Fe-N type II one. phenylahistin 62-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-47 18725511-7 2008 The highest contribution for the N-demethylation as calculated from the enzyme kinetic data, were obtained for CYP2B6 (R,S-MDMA), CYP1A2 (R-MDMA), and CYP2B6 (S-MDMA). Nitrogen 33-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 18725511-7 2008 The highest contribution for the N-demethylation as calculated from the enzyme kinetic data, were obtained for CYP2B6 (R,S-MDMA), CYP1A2 (R-MDMA), and CYP2B6 (S-MDMA). Nitrogen 33-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 18669587-0 2008 Generation of human metabolites of 7-ethoxycoumarin by bacterial cytochrome P450 BM3. 7-ethoxycoumarin 35-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 18669587-3 2008 In this report, we show that the oxidation of 7-ethoxycoumarin, a typical human P450 substrate, is catalyzed by both wild-type and mutant forms of CYP102A1. 7-ethoxycoumarin 46-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 18687804-10 2008 In contrast, (+)-phenylahistin mainly produced P3 in human microsomes and CYP3A human expressed P450s. phenylahistin 13-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-101 18687804-11 2008 (-)-Phenylahistin proved to be less toxic on P450-rich hepatocytes than on P450-deprived KB lines. phenylahistin 0-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-49 19211970-0 2008 Caffeine as a marker substrate for testing cytochrome P450 activity in human and rat. Caffeine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 19211970-1 2008 The current knowledge on the involvement of cytochrome P450 (P450, CYP) isoforms in the metabolism of caffeine in rat and human liver is reviewed. Caffeine 102-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 19211970-1 2008 The current knowledge on the involvement of cytochrome P450 (P450, CYP) isoforms in the metabolism of caffeine in rat and human liver is reviewed. Caffeine 102-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 19211970-3 2008 Finally, we discuss the P450-mediated metabolism of caffeine in relation to coffee addiction and drug interactions. Caffeine 52-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 19211970-4 2008 Due to its safety, favorable pharmacokinetic properties, and P450 isoform-selective metabolism, caffeine has great potential as a metabolic marker substance in both humans and rats, and as a more universal metabolic tool in the latter species. Caffeine 96-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 19211970-5 2008 However, the qualitative and relative quantitative contribution of P450 isoforms to the metabolism of caffeine is species- and concentration-dependent. Caffeine 102-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-71 18784455-12 2008 As HIV-infected hemodialysis patients are disproportionately black, the increased frequency of the CYP2B6 516G>T polymorphism may lead to higher EFV levels. efavirenz 148-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 18976212-1 2008 Cytochromes P450 represent a numerous family of heme-containing enzymes belonging to the group of monooxygenases. Heme 48-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-16 18729327-0 2008 Modification of serine 360 by a reactive intermediate of 17-alpha-ethynylestradiol results in mechanism-based inactivation of cytochrome P450s 2B1 and 2B6. Serine 16-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-154 18729327-0 2008 Modification of serine 360 by a reactive intermediate of 17-alpha-ethynylestradiol results in mechanism-based inactivation of cytochrome P450s 2B1 and 2B6. Ethinyl Estradiol 57-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-154 18600471-0 2008 Oxygen activation by cytochrome P450 monooxygenase. Oxygen 0-6 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Oxygen 61-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Oxygen 61-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Oxygen 183-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Oxygen 183-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Oxygen 185-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Oxygen 185-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Hydrocarbons 251-263 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Hydrocarbons 251-263 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Hydrogen 274-282 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Hydrogen 274-282 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Oxygen 222-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 18600471-1 2008 Unlike photosystem II (PSII) that catalyzes formation of the O-O bond, the cytochromes P450 (P450), members of a superfamily of hemoproteins, catalyze the scission of the O-O bond of dioxygen molecules and insert a single oxygen atom into unactivated hydrocarbons through a hydrogen abstraction-oxygen rebound mechanism. Oxygen 222-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 18600471-3 2008 Even though it carries out the opposite of the water splitting reaction, P450 may share similarities to PSII in proton delivery networks, oxygen and water access channels, and consecutive electron transfer processes. Water 47-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 18600471-3 2008 Even though it carries out the opposite of the water splitting reaction, P450 may share similarities to PSII in proton delivery networks, oxygen and water access channels, and consecutive electron transfer processes. Oxygen 138-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 18600471-3 2008 Even though it carries out the opposite of the water splitting reaction, P450 may share similarities to PSII in proton delivery networks, oxygen and water access channels, and consecutive electron transfer processes. Water 149-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 18600471-4 2008 In this article, we review recent advances in understanding the molecular mechanisms by which P450 activates dioxygen. Oxygen 109-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-98 18976212-6 2008 Under some conditions, cytochromes P450 can produce reactive oxygen species, and this is another problem attracting increasing attention. Reactive Oxygen Species 52-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 18474683-7 2008 OPZ also induced transcription of the human CYP2B6 promoter-reporter containing the phenobarbital (PB) responsive element in mouse liver using an in vivo transcription assay. Phenobarbital 84-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 18729332-5 2008 This is due to partitioning of the benzylic free radial intermediate between oxygen rebound to form 12-OH-NVP and loss of another hydrogen atom to form a reactive quinone methide, which inactivates P450. Oxygen 77-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-202 18729332-5 2008 This is due to partitioning of the benzylic free radial intermediate between oxygen rebound to form 12-OH-NVP and loss of another hydrogen atom to form a reactive quinone methide, which inactivates P450. Hydrogen 130-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-202 18729332-5 2008 This is due to partitioning of the benzylic free radial intermediate between oxygen rebound to form 12-OH-NVP and loss of another hydrogen atom to form a reactive quinone methide, which inactivates P450. quinone 163-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-202 18729332-6 2008 Cotreatment with the P450 inhibitor, 1-aminobenzotriazole, led to comparable levels of NVP and the deuterated analogue, and the deuterated analogue still caused a lower rash incidence. 1-aminobenzotriazole 37-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 18729332-8 2008 We propose that the hepatotoxicity of NVP in humans is due to the quinone methide formed by P450 in the liver, while the skin rash may be due to the quinone methide formed in the skin by sulfation of 12-OH metabolite followed by loss of sulfate. quinone 66-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 18611395-0 2008 The monoterpenoids citral and geraniol are moderate inhibitors of CYP2B6 hydroxylase activity. Monoterpenes 4-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 18611395-0 2008 The monoterpenoids citral and geraniol are moderate inhibitors of CYP2B6 hydroxylase activity. citral 19-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 18611395-0 2008 The monoterpenoids citral and geraniol are moderate inhibitors of CYP2B6 hydroxylase activity. geraniol 30-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 18611395-3 2008 We have recently discovered a moderate inhibitory effect of borneol and isoborneol toward CYP2B6-catalyzed bupropion hydroxylase activity. isoborneol 60-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 18611395-3 2008 We have recently discovered a moderate inhibitory effect of borneol and isoborneol toward CYP2B6-catalyzed bupropion hydroxylase activity. isoborneol 72-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 18611395-4 2008 Based on that result, we expanded our study to evaluate the inhibitory effects of 22 monoterpenoids on CYP2B6 activity in vitro. Monoterpenes 85-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 18611395-7 2008 These in vitro data indicate that high amounts of these two monoterpenoids might interact with drugs that are metabolized by CYP2B6. Monoterpenes 60-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 18611395-8 2008 The in vivo pharmacokinetics of these compounds should be examined to determine whether the inhibition of CYP2B6 activity by monoterpenoids has clinical relevance. Monoterpenes 125-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 18474675-0 2008 Cytochrome P450 2B6 catalyzes the formation of pharmacologically active sibutramine (N-{1-[1-(4-chlorophenyl)cyclobutyl]-3-methylbutyl}-N,N-dimethylamine) metabolites in human liver microsomes. sibutramine 72-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 18781911-5 2008 These individual differences may result in variable systemic exposure to drugs metabolized by CYP2B6, including the antineoplastics cyclophosphamide and ifosfamide, the antiretrovirals nevirapine and efavirenz, the anesthetics propofol and ketamine, the synthetic opioid methadone, and the anti-Parkinsonian selegiline. Cyclophosphamide 132-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 18781911-5 2008 These individual differences may result in variable systemic exposure to drugs metabolized by CYP2B6, including the antineoplastics cyclophosphamide and ifosfamide, the antiretrovirals nevirapine and efavirenz, the anesthetics propofol and ketamine, the synthetic opioid methadone, and the anti-Parkinsonian selegiline. Ifosfamide 153-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 18781911-5 2008 These individual differences may result in variable systemic exposure to drugs metabolized by CYP2B6, including the antineoplastics cyclophosphamide and ifosfamide, the antiretrovirals nevirapine and efavirenz, the anesthetics propofol and ketamine, the synthetic opioid methadone, and the anti-Parkinsonian selegiline. Nevirapine 185-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 18781911-5 2008 These individual differences may result in variable systemic exposure to drugs metabolized by CYP2B6, including the antineoplastics cyclophosphamide and ifosfamide, the antiretrovirals nevirapine and efavirenz, the anesthetics propofol and ketamine, the synthetic opioid methadone, and the anti-Parkinsonian selegiline. efavirenz 200-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 18781911-5 2008 These individual differences may result in variable systemic exposure to drugs metabolized by CYP2B6, including the antineoplastics cyclophosphamide and ifosfamide, the antiretrovirals nevirapine and efavirenz, the anesthetics propofol and ketamine, the synthetic opioid methadone, and the anti-Parkinsonian selegiline. Propofol 227-235 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 18781911-5 2008 These individual differences may result in variable systemic exposure to drugs metabolized by CYP2B6, including the antineoplastics cyclophosphamide and ifosfamide, the antiretrovirals nevirapine and efavirenz, the anesthetics propofol and ketamine, the synthetic opioid methadone, and the anti-Parkinsonian selegiline. Ketamine 240-248 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 18781911-5 2008 These individual differences may result in variable systemic exposure to drugs metabolized by CYP2B6, including the antineoplastics cyclophosphamide and ifosfamide, the antiretrovirals nevirapine and efavirenz, the anesthetics propofol and ketamine, the synthetic opioid methadone, and the anti-Parkinsonian selegiline. Methadone 271-280 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 18781911-5 2008 These individual differences may result in variable systemic exposure to drugs metabolized by CYP2B6, including the antineoplastics cyclophosphamide and ifosfamide, the antiretrovirals nevirapine and efavirenz, the anesthetics propofol and ketamine, the synthetic opioid methadone, and the anti-Parkinsonian selegiline. Selegiline 308-318 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 18728241-1 2008 The goal of this study was to determine the effect of CYP2B6 genetic variation on the steady-state pharmacokinetics of efavirenz (600 mg/d) in TB/HIV co-infected patients receiving concomitant rifampin, a potent CYP inducer. efavirenz 119-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 18728241-4 2008 Apparent oral clearance (CL/F) was significantly lower in patients with CYP2B6 c.516TT than in those with GT genotype (2.1 vs 8.4 mL/min/kg, P<0.0001) and GG genotype (2.1 vs 9.9 mL/min/kg, P< .0001). Fluorine 28-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 18728241-6 2008 Our results indicate that CYP2B6 c.516TT genotype can be used to identify efavirenz poor metabolizers in patients co-treated with rifampin. Rifampin 130-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 18474675-2 2008 Mechanism-based CYP2B6 inhibitors (i.e., clopidogrel, ticlopidine, and triethylenethiophoramide) significantly inhibited the formation of M1 from sibutramine and M2 from M1, respectively; in contrast, no effect was observed when using potent inhibitors of eight P450 isozymes (CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A). Ticlopidine 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 18474675-2 2008 Mechanism-based CYP2B6 inhibitors (i.e., clopidogrel, ticlopidine, and triethylenethiophoramide) significantly inhibited the formation of M1 from sibutramine and M2 from M1, respectively; in contrast, no effect was observed when using potent inhibitors of eight P450 isozymes (CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A). triethylenethiophoramide 71-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 18474675-2 2008 Mechanism-based CYP2B6 inhibitors (i.e., clopidogrel, ticlopidine, and triethylenethiophoramide) significantly inhibited the formation of M1 from sibutramine and M2 from M1, respectively; in contrast, no effect was observed when using potent inhibitors of eight P450 isozymes (CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A). sibutramine 146-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 18474675-3 2008 In addition, the formations of M1 from sibutramine (r = 0.694, p = 0.0029) and M2 from M1 (r = 0.834, p < 0.0001) were strongly correlated with CYP2B6-catalyzed bupropion hydroxylation in 16 different HLM panels. sibutramine 39-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 18474675-3 2008 In addition, the formations of M1 from sibutramine (r = 0.694, p = 0.0029) and M2 from M1 (r = 0.834, p < 0.0001) were strongly correlated with CYP2B6-catalyzed bupropion hydroxylation in 16 different HLM panels. Bupropion 164-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 18474675-7 2008 In conclusion, CYP2B6 is the primary catalyst for the formation of sibutramine two active metabolites, which may suggest that pharmacogenetics and drug interactions of sibutramine in relation to CYP2B6 activity should be considered in the pharmacotherapy of sibutramine. sibutramine 67-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 18474675-7 2008 In conclusion, CYP2B6 is the primary catalyst for the formation of sibutramine two active metabolites, which may suggest that pharmacogenetics and drug interactions of sibutramine in relation to CYP2B6 activity should be considered in the pharmacotherapy of sibutramine. sibutramine 67-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-201 18474675-7 2008 In conclusion, CYP2B6 is the primary catalyst for the formation of sibutramine two active metabolites, which may suggest that pharmacogenetics and drug interactions of sibutramine in relation to CYP2B6 activity should be considered in the pharmacotherapy of sibutramine. sibutramine 168-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 18474675-0 2008 Cytochrome P450 2B6 catalyzes the formation of pharmacologically active sibutramine (N-{1-[1-(4-chlorophenyl)cyclobutyl]-3-methylbutyl}-N,N-dimethylamine) metabolites in human liver microsomes. n-{1-[1-(4-chlorophenyl)cyclobutyl]-3-methylbutyl}-n,n-dimethylamine 85-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 18474675-2 2008 Mechanism-based CYP2B6 inhibitors (i.e., clopidogrel, ticlopidine, and triethylenethiophoramide) significantly inhibited the formation of M1 from sibutramine and M2 from M1, respectively; in contrast, no effect was observed when using potent inhibitors of eight P450 isozymes (CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A). Clopidogrel 41-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 18474675-7 2008 In conclusion, CYP2B6 is the primary catalyst for the formation of sibutramine two active metabolites, which may suggest that pharmacogenetics and drug interactions of sibutramine in relation to CYP2B6 activity should be considered in the pharmacotherapy of sibutramine. sibutramine 168-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 18474683-7 2008 OPZ also induced transcription of the human CYP2B6 promoter-reporter containing the phenobarbital (PB) responsive element in mouse liver using an in vivo transcription assay. Phenobarbital 99-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 18480186-3 2008 The curcuminoid extract inhibited SULT > CYP2C19 > CYP2B6 > UGT > CYP2C9 > CYP3A activities with IC(50) values ranging from 0.99 +/- 0.04 to 25.3 +/- 1.3 microM, whereas CYP2D6, CYP1A2, and CYP2E1 activities were less affected (IC(50) values > 60 microM). curcuminoid 4-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 18433425-0 2008 Relationship of cytochrome P450 pharmacogenetics to the effects of yohimbine on gastrointestinal transit and catecholamines in healthy subjects. Yohimbine 67-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 18433425-7 2008 As cytochrome P450 enzymes metabolize yohimbine, P450 genotypes (CYP2D6 and CYP3A4) were determined in 25 of 30 subjects who consented to genetic studies. Yohimbine 38-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 18372400-10 2008 The results from in vitro experiments with recombinant isoforms and P450 isoform-selective inhibitors suggested that the oxidative metabolism of lasofoxifene is catalyzed primarily by CYP3A and CYP2D6. Lasofoxifene 145-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 18385292-8 2008 The P450 probe substrates indicate a decrease in CYP1A2, CYP2B6, CYP2C9, and CYP3A4 activities in HepaRG cells 1 day after removal of DMSO from the medium. Dimethyl Sulfoxide 134-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-8 18385292-2 2008 The expression of cytochrome P450 (P450) enzymes, transporter proteins, and transcription factors was stable in differentiated HepaRG cells over a period of 6 weeks when cultured with DMSO. Dimethyl Sulfoxide 184-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 18385292-8 2008 The P450 probe substrates indicate a decrease in CYP1A2, CYP2B6, CYP2C9, and CYP3A4 activities in HepaRG cells 1 day after removal of DMSO from the medium. Dimethyl Sulfoxide 134-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 18385292-2 2008 The expression of cytochrome P450 (P450) enzymes, transporter proteins, and transcription factors was stable in differentiated HepaRG cells over a period of 6 weeks when cultured with DMSO. Dimethyl Sulfoxide 184-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 18673154-0 2008 Prediction of cytochrome P450 2B6-substrate interactions using pharmacophore ensemble/support vector machine (PhE/SVM) approach. Phenylalanine 110-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-33 18496131-10 2008 CONCLUSION: This study indicates that the presently evaluated variant alleles in the CYP2B6, CYP2C9, CYP2C19, CYP3A4, CYP3A5, GSTA1, GSTP1, ALDH1A1 and ALDH3A1 genes do not explain the interindividual variability in cyclophosphamide and 4-hydroxycyclophosphamide pharmacokinetics and are, probably, not the cause of the observed variability in toxicity. Cyclophosphamide 216-232 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 18332078-8 2008 CYP3A4 and CYP2B6 induction in Fa2N-4 cells were also low for phenytoin, phenobarbital, and efavirenz, which are dual activators of PXR/CAR. Phenytoin 62-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 18332078-8 2008 CYP3A4 and CYP2B6 induction in Fa2N-4 cells were also low for phenytoin, phenobarbital, and efavirenz, which are dual activators of PXR/CAR. Phenobarbital 73-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 18332078-8 2008 CYP3A4 and CYP2B6 induction in Fa2N-4 cells were also low for phenytoin, phenobarbital, and efavirenz, which are dual activators of PXR/CAR. efavirenz 92-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 18332082-4 2008 To identify individual constituents with inhibitory activity, the suspension was partitioned using hexane, ethyl acetate, and dichloromethane, and each fraction was tested for its inhibitory effect on CYP2B6-catalyzed bupropion hydroxylation. Bupropion 218-227 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 201-207 18332082-6 2008 These two terpenoids moderately inhibited CYP2B6-catalyzed bupropion hydroxylase activity in a competitive manner, with K(i) values of 9.5 and 5.9 microM, respectively, as well as efavirenz 8-hydroxylase activity, with K(i) values of 22 and 26 microM, respectively. Terpenes 10-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 18478283-3 2008 In this work, the five structurally modified heme derivatives 2-6 and the native heme 1 were docked on CYP2B4, (an isoform of P450), in order to determine whether such modifications alter their binding form and binding affinity for CYP2B4 apoprotein. Heme 45-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 18478283-3 2008 In this work, the five structurally modified heme derivatives 2-6 and the native heme 1 were docked on CYP2B4, (an isoform of P450), in order to determine whether such modifications alter their binding form and binding affinity for CYP2B4 apoprotein. Heme 81-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 18219560-0 2008 Stereoselective metabolism of bupropion by cytochrome P4502B6 (CYP2B6) and human liver microsomes. Bupropion 30-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 18219560-2 2008 Bupropion hydroxylation is catalyzed selectively by cytochrome P4502B6 (CYP2B6). Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 18219560-3 2008 CYP2B6-catalyzed bupropion hydroxylation has been used as an in vitro and in vivo phenotypic probe for CYP2B6 activity and CYP2B6 drug interactions. Bupropion 17-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 18219560-3 2008 CYP2B6-catalyzed bupropion hydroxylation has been used as an in vitro and in vivo phenotypic probe for CYP2B6 activity and CYP2B6 drug interactions. Bupropion 17-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 18219560-3 2008 CYP2B6-catalyzed bupropion hydroxylation has been used as an in vitro and in vivo phenotypic probe for CYP2B6 activity and CYP2B6 drug interactions. Bupropion 17-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 18496131-10 2008 CONCLUSION: This study indicates that the presently evaluated variant alleles in the CYP2B6, CYP2C9, CYP2C19, CYP3A4, CYP3A5, GSTA1, GSTP1, ALDH1A1 and ALDH3A1 genes do not explain the interindividual variability in cyclophosphamide and 4-hydroxycyclophosphamide pharmacokinetics and are, probably, not the cause of the observed variability in toxicity. hydroxycyclophosphamide 239-262 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 18570159-4 2008 Treatment of human hepatocytes for 72 h with 2-200 microM TB produced concentration-dependent increases in CYP1A2, CYP2B6 and CYP3A4 mRNA levels, whereas treatment with BHT increased CYP2B6 and CYP3A4 mRNA levels. Thiabendazole 58-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 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. Butylated Hydroxytoluene 144-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-170 18570159-4 2008 Treatment of human hepatocytes for 72 h with 2-200 microM TB produced concentration-dependent increases in CYP1A2, CYP2B6 and CYP3A4 mRNA levels, whereas treatment with BHT increased CYP2B6 and CYP3A4 mRNA levels. Thiabendazole 58-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-189 18570159-4 2008 Treatment of human hepatocytes for 72 h with 2-200 microM TB produced concentration-dependent increases in CYP1A2, CYP2B6 and CYP3A4 mRNA levels, whereas treatment with BHT increased CYP2B6 and CYP3A4 mRNA levels. Butylated Hydroxytoluene 169-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-189 18570159-5 2008 CYP1A2, CYP2B6 and CYP3A4 mRNA levels were induced around 48-, 21- and 9-fold, respectively, by 200 microM TB, with CYP2B6 and CYP 3A4 mRNA levels being induced around 12- and 7-fold, respectively, by 200 microM BHT. Thiabendazole 107-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 18570159-5 2008 CYP1A2, CYP2B6 and CYP3A4 mRNA levels were induced around 48-, 21- and 9-fold, respectively, by 200 microM TB, with CYP2B6 and CYP 3A4 mRNA levels being induced around 12- and 7-fold, respectively, by 200 microM BHT. Thiabendazole 107-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 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 2 subfamily B member 6 Homo sapiens 113-118 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 2 subfamily B member 6 Homo sapiens 165-170 18057928-9 2008 Pharmacokinetic parameter estimates indicate that a dose reduction to 400 mg efavirenz per day is possible in patients homozygous for the CYP2B6*6 genotype without compromising therapeutic efficacy. efavirenz 77-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 18285471-4 2008 Ritonavir induces CYP2B6 in human hepatocytes. Ritonavir 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 18285471-5 2008 This investigation tested the hypothesis that ritonavir induces human CYP2B6 in vivo. Ritonavir 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 18285471-7 2008 Stereoselective bupropion hydroxylation was used as an in vivo probe for CYP2B6 activity. Bupropion 16-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 18285471-13 2008 These results show that ritonavir induces human CYP2B6 activity. Ritonavir 24-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-54 18285471-15 2008 The ritonavir induction of CYP2B6 activity may have significant implications for drug interactions and clarify previously unexplained interactions. Ritonavir 4-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 18331760-0 2008 Reversible inactivation of cytochrome P450 by alkaline earth metal ions: auxiliary metal ion induced conformation change and formation of inactive P420 species in CYP101. Metals 61-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 18331760-0 2008 Reversible inactivation of cytochrome P450 by alkaline earth metal ions: auxiliary metal ion induced conformation change and formation of inactive P420 species in CYP101. Metals 83-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 18331760-8 2008 The results have been discussed in the light of understanding the mechanism of inactivation of certain mammalian P450 enzymes by these alkaline-earth metal ions. Metals 150-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-117 18164676-4 2008 In an anaerobic solution, direct and reversible electron transfer between the electroactive heme center of CYP2B6 and the electrode was observed with a formal potential of -0.454 +/- 0.006 V at pH 7.4. Heme 92-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 18219560-5 2008 Nevertheless, it is unknown whether CYP2B6-catalyzed bupropion hydroxylation is stereoselective. Bupropion 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 18219560-6 2008 METHODS: Hydroxylation of racemic bupropion by recombinant CYP2B6 and human liver microsomes was evaluated using a stereoselective assay. Bupropion 34-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 18219560-7 2008 RESULTS: At therapeutic concentrations, hydroxylation of (S)-bupropion was threefold and 1.5-greater than (R)-bupropion, respectively, by recombinant CYP2B6 and human liver microsomes. Bupropion 57-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 18219560-7 2008 RESULTS: At therapeutic concentrations, hydroxylation of (S)-bupropion was threefold and 1.5-greater than (R)-bupropion, respectively, by recombinant CYP2B6 and human liver microsomes. (R)-bupropion 106-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 18219560-9 2008 CONCLUSIONS: Stereoselective bupropion hydroxylation may have implications for the therapeutic efficacy of bupropion as an antidepressant or smoking cessation therapy, and for the use of bupropion as an in vivo phenotypic probe for CYP2B6 activity. Bupropion 29-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 232-238 18071681-0 2008 Assessment of urinary mephenytoin metrics to phenotype for CYP2C19 and CYP2B6 activity. Mephenytoin 22-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 18071681-4 2008 We also assessed whether urinary excretion of N-desmethylmephenytoin (nirvanol) might be a useful CYP2B6 metric in in vivo studies. ethylphenylhydantoin 70-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 18071681-11 2008 While nirvanol excretion may reflect CYP2B6 activity in vivo, it is not useful for CYP2B6 phenotyping. ethylphenylhydantoin 6-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 18350255-5 2008 The production rate of CYP2B6 was increased 79.7% by artemisinin, 61.5% by arteether, 76.1% by artemether, 19.9% by dihydroartemisinin and 16.9% by artesunate. Artesunate 148-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 18281305-9 2008 CONCLUSIONS: This is the first report that the 983T>C genotype (part of the CYP2B6*18 haplotype) impacts on nevirapine plasma concentrations and the first study to assess the impact of 983C homozygosity on efavirenz concentrations. Nevirapine 111-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 18287571-0 2008 Stereoselective bupropion hydroxylation as an in vivo phenotypic probe for cytochrome P4502B6 (CYP2B6) activity. Bupropion 16-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-101 18287571-1 2008 The clearance of racemic bupropion, metabolized selectively by CYP2B6 in vitro, has been used clinically to phenotype CYP2B6 activity, polymorphisms, and drug interactions but has known limitations. Bupropion 25-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 18287571-1 2008 The clearance of racemic bupropion, metabolized selectively by CYP2B6 in vitro, has been used clinically to phenotype CYP2B6 activity, polymorphisms, and drug interactions but has known limitations. Bupropion 25-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 18287571-2 2008 Bupropion hydroxylation by CYP2B6 is stereoselective. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 18171905-1 2008 CYP2B6 is a polymorphic human drug metabolizing cytochrome P450 with clinical relevance for several drug substrates including cyclophosphamide, bupropion, and efavirenz. Cyclophosphamide 126-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 18171905-1 2008 CYP2B6 is a polymorphic human drug metabolizing cytochrome P450 with clinical relevance for several drug substrates including cyclophosphamide, bupropion, and efavirenz. Bupropion 144-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 18171905-1 2008 CYP2B6 is a polymorphic human drug metabolizing cytochrome P450 with clinical relevance for several drug substrates including cyclophosphamide, bupropion, and efavirenz. efavirenz 159-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 18350255-0 2008 A model based assessment of the CYP2B6 and CYP2C19 inductive properties by artemisinin antimalarials: implications for combination regimens. artemisinin 75-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 18350255-1 2008 The study aim was to assess the inductive properties of artemisinin antimalarials using mephenytoin as a probe for CYP2B6 and CYP2C19 enzymatic activity. artemisinin 56-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 18350255-1 2008 The study aim was to assess the inductive properties of artemisinin antimalarials using mephenytoin as a probe for CYP2B6 and CYP2C19 enzymatic activity. Mephenytoin 88-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 18350255-5 2008 The production rate of CYP2B6 was increased 79.7% by artemisinin, 61.5% by arteether, 76.1% by artemether, 19.9% by dihydroartemisinin and 16.9% by artesunate. artemisinin 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 18350255-5 2008 The production rate of CYP2B6 was increased 79.7% by artemisinin, 61.5% by arteether, 76.1% by artemether, 19.9% by dihydroartemisinin and 16.9% by artesunate. artemotil 75-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 18350255-5 2008 The production rate of CYP2B6 was increased 79.7% by artemisinin, 61.5% by arteether, 76.1% by artemether, 19.9% by dihydroartemisinin and 16.9% by artesunate. Artemether 95-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 18350255-5 2008 The production rate of CYP2B6 was increased 79.7% by artemisinin, 61.5% by arteether, 76.1% by artemether, 19.9% by dihydroartemisinin and 16.9% by artesunate. artenimol 116-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 18171905-6 2008 Further investigation using minigene constructs transfected into eukaryotic cell lines COS-1 and Huh7 demonstrated that the single nucleotide polymorphism c.516G>T in allele CYP2B6*6 was alone responsible for aberrant splicing resulting in high-splice variant (SV) 1 and low-CYP2B6 expression phenotype. carbonyl sulfide 87-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-183 18171905-6 2008 Further investigation using minigene constructs transfected into eukaryotic cell lines COS-1 and Huh7 demonstrated that the single nucleotide polymorphism c.516G>T in allele CYP2B6*6 was alone responsible for aberrant splicing resulting in high-splice variant (SV) 1 and low-CYP2B6 expression phenotype. carbonyl sulfide 87-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 278-284 18350255-7 2008 In conclusion, all studied artemisinin derivatives induced CYP2B6. artemisinin 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 19326768-9 2008 At 100 microM deltamethrin 2-3 fold induction of CYP1A1 and CYP2B6 mRNA was observed, while at the same time an approximately 25-fold induction of CYP3A4 was noted. decamethrin 14-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 18096694-5 2008 We found that coumestrol was able to suppress the effects of PXR agonists on the expression of the known PXR target genes, CYP3A4 and CYP2B6, in primary human hepatocytes as well as inhibit metabolism of tribromoethanol in humanized PXR mice. Coumestrol 14-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-140 18292673-10 2008 RESULTS: At clinical concentrations, methadone enantiomer N-demethylation by recombinant CYPs 2B6, 3A4, and 2C19 was S > R, S = R, and S << R. Greater stereoselective metabolism (S > R) occurred in livers expressing high levels of CYP2B6 compared with CYP3A4. Nitrogen 58-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 243-249 18292673-15 2008 CONCLUSIONS: These results suggest a significant role for CYP2B6, but not CYP3A, in stereoselective human methadone metabolism and disposition. Methadone 106-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 18094037-11 2008 Phenobarbital (constitutive androstane receptor activator) induced CYP3A4 (4.1-fold, only in jejunum), CYP2B6 (4.9-fold in colon and 2.3-fold in proximal jejunum), and MDR1/ABCB1 mRNA and CYP3A4 activity (2-fold only proximal jejunum). Phenobarbital 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 18642140-0 2008 Mechanism and role of covalent heme binding in the CYP4 family of P450 enzymes and the mammalian peroxidases. Heme 31-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 18642140-1 2008 The CYP4 family of cytochrome P450 enzymes and the mammalian peroxidases covalently bind their heme groups. Heme 95-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 18212249-0 2008 Improvement of cyclophosphamide activation by CYP2B6 mutants: from in silico to ex vivo. Cyclophosphamide 15-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 18212249-1 2008 Cyclophosphamide (CPA) is a chemotherapeutic agent that is primarily activated in the liver by cytochrome P4502B6 (CYP2B6) and then transported to the tumor via blood flow. Cyclophosphamide 0-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 18212249-1 2008 Cyclophosphamide (CPA) is a chemotherapeutic agent that is primarily activated in the liver by cytochrome P4502B6 (CYP2B6) and then transported to the tumor via blood flow. Cyclophosphamide 18-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 18212249-3 2008 Given the relatively low affinity of CYP2B6 for CPA, the aim of our work was to modify CYP2B6 to increase its catalytic efficiency (V(max)/K(m)) to metabolize CPA into 4"-OH CPA. Cyclophosphamide 48-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 18212249-3 2008 Given the relatively low affinity of CYP2B6 for CPA, the aim of our work was to modify CYP2B6 to increase its catalytic efficiency (V(max)/K(m)) to metabolize CPA into 4"-OH CPA. Cyclophosphamide 48-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 18212249-3 2008 Given the relatively low affinity of CYP2B6 for CPA, the aim of our work was to modify CYP2B6 to increase its catalytic efficiency (V(max)/K(m)) to metabolize CPA into 4"-OH CPA. Cyclophosphamide 159-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 18212249-3 2008 Given the relatively low affinity of CYP2B6 for CPA, the aim of our work was to modify CYP2B6 to increase its catalytic efficiency (V(max)/K(m)) to metabolize CPA into 4"-OH CPA. Cyclophosphamide 159-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 18212249-3 2008 Given the relatively low affinity of CYP2B6 for CPA, the aim of our work was to modify CYP2B6 to increase its catalytic efficiency (V(max)/K(m)) to metabolize CPA into 4"-OH CPA. 4"-oh cpa 168-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 18212249-3 2008 Given the relatively low affinity of CYP2B6 for CPA, the aim of our work was to modify CYP2B6 to increase its catalytic efficiency (V(max)/K(m)) to metabolize CPA into 4"-OH CPA. 4"-oh cpa 168-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 18212249-5 2008 Canine CYP2B11 exhibiting a particularly low K(m) to CPA, the amino acids exclusively present in the CYP2B11 substrate recognition sequences were substituted in human CYP2B6. Cyclophosphamide 53-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-173 18212249-10 2008 Finally, expression of the CYP2B6 114V/477W double mutant, contrary to wt CYP2B6, allowed switching of a resistant human head and neck cancer cell line (A-253) into a sensitive cell line toward CPA. Cyclophosphamide 194-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 18212249-11 2008 Thus, we were able to obtain a new efficient CYP2B6 mutant able to metabolize CPA, an important step in the GDEPT strategy for human cancer treatment. Cyclophosphamide 78-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 18206661-3 2008 To ensure high-level expression of CYP2B6, the inducer phenytoin was present at all times. Phenytoin 55-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 18206661-6 2008 Nitric oxide synthase inhibitors attenuated the downregulation of CYP2B6 protein but not mRNA by ILmix. Nitric Oxide 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 18292673-0 2008 Role of CYP2B6 in stereoselective human methadone metabolism. Methadone 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 18292673-4 2008 Nevertheless, recent clinical data do not support a primary role for CYP3A4 and suggest that CYP2B6 may mediate methadone clearance. Methadone 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 18292673-7 2008 METHODS: N-demethylation of racemic methadone and individual enantiomers by expressed CYPs 2B6, 2C19, and 3A4 was evaluated. Nitrogen 9-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-100 18292673-7 2008 METHODS: N-demethylation of racemic methadone and individual enantiomers by expressed CYPs 2B6, 2C19, and 3A4 was evaluated. Methadone 36-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-100 18292673-10 2008 RESULTS: At clinical concentrations, methadone enantiomer N-demethylation by recombinant CYPs 2B6, 3A4, and 2C19 was S > R, S = R, and S << R. Greater stereoselective metabolism (S > R) occurred in livers expressing high levels of CYP2B6 compared with CYP3A4. Methadone 37-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 243-249 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 21-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 18094037-8 2008 In proximal jejunum, rifampicin (RIF) [pregnane X receptor (PXR) ligand] induced CYP3A4 (5.2-fold), CYP2B6 (2-fold), UGT1A6 (2.2-fold), and multidrug resistance-1 (MDR1)/ABCB1 mRNA (2.7-fold), whereas 6beta-hydroxytestosterone formation (CYP3A4) increased 2-fold. Rifampin 33-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 17875191-0 2008 Serious haematological toxicity of cyclophosphamide in relation to CYP2B6, GSTA1 and GSTP1 polymorphisms. Cyclophosphamide 35-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 18161730-1 2008 The hydroxylation activity of the Thr268Ala mutant of P450(BM3) has been shown to occur to varying degrees with small alterations in the structure of a fatty-acid substrate. Fatty Acids 152-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-62 18022748-6 2008 In human hepatocytes Pyrethrins and Phenobarbital induced both testosterone 6beta-hydroxylase activity and CYP3A4 mRNA levels and also increased CYP2B6 mRNA levels. Pyrethrins 21-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-151 18022748-6 2008 In human hepatocytes Pyrethrins and Phenobarbital induced both testosterone 6beta-hydroxylase activity and CYP3A4 mRNA levels and also increased CYP2B6 mRNA levels. Phenobarbital 36-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-151 19122335-2 2008 Ticlopidine and clopidogrel inhibited CYP2B6 with IC(50) values of 0.0517+/-0.0323 microM and 0.0182+/-0.0069 microM, respectively, and inhibited CYP2C19 with IC(50) values of 0.203+/-0.124 microM and 0.524+/-0.160 microM, respectively. Ticlopidine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 19122335-2 2008 Ticlopidine and clopidogrel inhibited CYP2B6 with IC(50) values of 0.0517+/-0.0323 microM and 0.0182+/-0.0069 microM, respectively, and inhibited CYP2C19 with IC(50) values of 0.203+/-0.124 microM and 0.524+/-0.160 microM, respectively. Clopidogrel 16-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 19122335-4 2008 In contrast, 2-oxo-clopidogrel, prasugrel and R-95913 were much weaker inhibitors of CYP2B6, CYP2C19 and CYP2D6. 2-oxo-clopidogrel 13-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 19122335-4 2008 In contrast, 2-oxo-clopidogrel, prasugrel and R-95913 were much weaker inhibitors of CYP2B6, CYP2C19 and CYP2D6. Prasugrel Hydrochloride 32-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 19122335-4 2008 In contrast, 2-oxo-clopidogrel, prasugrel and R-95913 were much weaker inhibitors of CYP2B6, CYP2C19 and CYP2D6. R-95913 46-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 19326768-10 2008 Permethrin-mediated CYP induction was much less potent, 4-fold or less for CYP1A1, CYP3A4, CYP3A5, CYP2B6 and CYP2A6. Permethrin 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 18493130-10 2008 The discrimination between 17alpha-hydroxylase and 17,20 lyase activities is regulated by two posttranslational events, the serine phosphorylation of P450c17 and the allosteric action of cytochrome b5, both of which act to optimize the interaction of P450c17 with its obligatory electron donor, P450 oxidoreductase. Serine 124-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 19326769-4 2008 DEET significantly induced CYP3A4, CYP2B6, CYP2A6 and CYP1A2 mRNA expression while chlorpyrifos induced CYP1A1, CYP1A2 and CYP3A4 mRNA, and to a lesser extent, CYP1B1 and CYP2B6 mRNA in primary human hepatocytes. Chlorpyrifos 83-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 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 2 subfamily B member 6 Homo sapiens 89-95 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 2 subfamily B member 6 Homo sapiens 89-95 21197270-5 2008 Other studies have shown associations of the CYP2B6-G516T genotype with nevirapine pharmacokinetics and central nervous system adverse effects related to efavirenz use. Nevirapine 72-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 18094219-2 2008 This open-label, multiple-dose, 2-period, fixed-sequence study assessed CYP2B6 inhibition by prasugrel using bupropion as a probe substrate, where its active metabolite, hydroxybupropion, is almost exclusively formed by CYP2B6. Prasugrel Hydrochloride 93-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 18094219-2 2008 This open-label, multiple-dose, 2-period, fixed-sequence study assessed CYP2B6 inhibition by prasugrel using bupropion as a probe substrate, where its active metabolite, hydroxybupropion, is almost exclusively formed by CYP2B6. hydroxybupropion 170-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 18094219-2 2008 This open-label, multiple-dose, 2-period, fixed-sequence study assessed CYP2B6 inhibition by prasugrel using bupropion as a probe substrate, where its active metabolite, hydroxybupropion, is almost exclusively formed by CYP2B6. hydroxybupropion 170-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-226 18094219-6 2008 Prasugrel weakly inhibited CYP2B6 activity as it increased bupropion Cmax and AUC0-infinity by 14% and 18%, respectively, and decreased hydroxybupropion Cmax and AUC0-infinity by 32% and 23%. Prasugrel Hydrochloride 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 18094219-6 2008 Prasugrel weakly inhibited CYP2B6 activity as it increased bupropion Cmax and AUC0-infinity by 14% and 18%, respectively, and decreased hydroxybupropion Cmax and AUC0-infinity by 32% and 23%. Bupropion 59-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 18094219-6 2008 Prasugrel weakly inhibited CYP2B6 activity as it increased bupropion Cmax and AUC0-infinity by 14% and 18%, respectively, and decreased hydroxybupropion Cmax and AUC0-infinity by 32% and 23%. hydroxybupropion cmax 136-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 18094219-7 2008 These results are consistent with patients receiving prasugrel not requiring dose adjustments when treated with drugs primarily metabolized by CYP2B6. Prasugrel Hydrochloride 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 18094219-0 2008 Prasugrel, a new thienopyridine antiplatelet drug, weakly inhibits cytochrome P450 2B6 in humans. Prasugrel Hydrochloride 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-86 17904175-2 2007 Phenobarbital (PB) induction of human CYP 2B6 and mouse CYP 2b10 has been shown to be mediated by CAR. Phenobarbital 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-45 17954932-0 2007 Regulation of iron homeostasis mediated by the heme-binding protein Dap1 (damage resistance protein 1) via the P450 protein Erg11/Cyp51. Iron 14-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-115 17954932-2 2007 In particular, the P450 protein Erg11/Cyp51 catalyzes a critical step in ergosterol synthesis, and the azole class of antifungal drugs inhibits Erg11. Ergosterol 73-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 17954932-2 2007 In particular, the P450 protein Erg11/Cyp51 catalyzes a critical step in ergosterol synthesis, and the azole class of antifungal drugs inhibits Erg11. Azoles 103-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 17954932-9 2007 The results suggest that Saccharomyces cerevisiae Dap1 stimulates a P450-catalyzed step in sterol synthesis via a distinct localization from its homologues in Schizosaccharomyces pombe and mammals and that this function regulates iron metabolism. Sterols 91-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 17954932-9 2007 The results suggest that Saccharomyces cerevisiae Dap1 stimulates a P450-catalyzed step in sterol synthesis via a distinct localization from its homologues in Schizosaccharomyces pombe and mammals and that this function regulates iron metabolism. Iron 230-234 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 17904175-2 2007 Phenobarbital (PB) induction of human CYP 2B6 and mouse CYP 2b10 has been shown to be mediated by CAR. Phenobarbital 15-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-45 17918089-8 2007 All EFV-treated CYP2B6 *6/*6 and *6/*26 carriers had extremely high plasma EFV concentrations (>6000 ng/mL) while receiving the standard dosage. efavirenz 4-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 17918089-8 2007 All EFV-treated CYP2B6 *6/*6 and *6/*26 carriers had extremely high plasma EFV concentrations (>6000 ng/mL) while receiving the standard dosage. efavirenz 75-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 17918089-1 2007 BACKGROUND: Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6), and high plasma concentrations of the drug are associated with a G-->T polymorphism at position 516 (516G-->T) of CYP2B6 and frequent central nervous system (CNS)-related side effects. efavirenz 12-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-75 17918089-1 2007 BACKGROUND: Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6), and high plasma concentrations of the drug are associated with a G-->T polymorphism at position 516 (516G-->T) of CYP2B6 and frequent central nervous system (CNS)-related side effects. efavirenz 12-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 17918089-10 2007 EFV-containing treatment was initiated at 400 mg in 4 CYP2B6 *6/*6 carriers and one *6/*26 carrier. efavirenz 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 17918089-1 2007 BACKGROUND: Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6), and high plasma concentrations of the drug are associated with a G-->T polymorphism at position 516 (516G-->T) of CYP2B6 and frequent central nervous system (CNS)-related side effects. efavirenz 12-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 206-212 17918089-13 2007 CONCLUSIONS: Genotype-based EFV dose reduction is feasible in CYP2B6 *6/*6 and *6/*26 carriers, which can reduce EFV-associated CNS symptoms. efavirenz 28-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 17918089-1 2007 BACKGROUND: Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6), and high plasma concentrations of the drug are associated with a G-->T polymorphism at position 516 (516G-->T) of CYP2B6 and frequent central nervous system (CNS)-related side effects. efavirenz 23-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-75 17918089-1 2007 BACKGROUND: Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6), and high plasma concentrations of the drug are associated with a G-->T polymorphism at position 516 (516G-->T) of CYP2B6 and frequent central nervous system (CNS)-related side effects. efavirenz 23-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 17918089-13 2007 CONCLUSIONS: Genotype-based EFV dose reduction is feasible in CYP2B6 *6/*6 and *6/*26 carriers, which can reduce EFV-associated CNS symptoms. efavirenz 113-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 17918089-1 2007 BACKGROUND: Efavirenz (EFV) is metabolized primarily by cytochrome P450 2B6 (CYP2B6), and high plasma concentrations of the drug are associated with a G-->T polymorphism at position 516 (516G-->T) of CYP2B6 and frequent central nervous system (CNS)-related side effects. efavirenz 23-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 206-212 17682072-3 2007 In this report, we describe the selectivity of 2-phenyl-2-(1-piperidinyl)propane (PPP) as an inactivator of CYP2B6 and compare this selectivity versus other CYP2B6 inactivators: 1,1",1""-phosphinothioylidynetrisaziridine (thioTEPA), clopidogrel, and ticlopidine. 2-phenyl-2-(1-piperidinyl)propane 47-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 17918089-4 2007 EFV dose was reduced in CYP2B6 516G-->T carriers who had high plasma EFV concentrations while receiving the standard dosage (600 mg). efavirenz 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 17682072-3 2007 In this report, we describe the selectivity of 2-phenyl-2-(1-piperidinyl)propane (PPP) as an inactivator of CYP2B6 and compare this selectivity versus other CYP2B6 inactivators: 1,1",1""-phosphinothioylidynetrisaziridine (thioTEPA), clopidogrel, and ticlopidine. 2-phenyl-2-(1-piperidinyl)propane 82-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 17682072-3 2007 In this report, we describe the selectivity of 2-phenyl-2-(1-piperidinyl)propane (PPP) as an inactivator of CYP2B6 and compare this selectivity versus other CYP2B6 inactivators: 1,1",1""-phosphinothioylidynetrisaziridine (thioTEPA), clopidogrel, and ticlopidine. Clopidogrel 233-244 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 17682072-3 2007 In this report, we describe the selectivity of 2-phenyl-2-(1-piperidinyl)propane (PPP) as an inactivator of CYP2B6 and compare this selectivity versus other CYP2B6 inactivators: 1,1",1""-phosphinothioylidynetrisaziridine (thioTEPA), clopidogrel, and ticlopidine. Thiotepa 178-220 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 17682072-3 2007 In this report, we describe the selectivity of 2-phenyl-2-(1-piperidinyl)propane (PPP) as an inactivator of CYP2B6 and compare this selectivity versus other CYP2B6 inactivators: 1,1",1""-phosphinothioylidynetrisaziridine (thioTEPA), clopidogrel, and ticlopidine. Ticlopidine 250-261 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 17682072-3 2007 In this report, we describe the selectivity of 2-phenyl-2-(1-piperidinyl)propane (PPP) as an inactivator of CYP2B6 and compare this selectivity versus other CYP2B6 inactivators: 1,1",1""-phosphinothioylidynetrisaziridine (thioTEPA), clopidogrel, and ticlopidine. Thiotepa 222-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 17979764-0 2007 Functional regulation of hepatic cytochrome p450 enzymes by physicochemical properties of phospholipids in biological membranes. Phospholipids 90-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 17709370-5 2007 Acetonitrile, DMSO, and ethanol were shown to increase the Km and decrease the intrinsic clearance (CLint) of CYP2B6-mediated bupropion hydroxylation in a concentration-dependent manner. acetonitrile 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 17709370-5 2007 Acetonitrile, DMSO, and ethanol were shown to increase the Km and decrease the intrinsic clearance (CLint) of CYP2B6-mediated bupropion hydroxylation in a concentration-dependent manner. Dimethyl Sulfoxide 14-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 17709370-5 2007 Acetonitrile, DMSO, and ethanol were shown to increase the Km and decrease the intrinsic clearance (CLint) of CYP2B6-mediated bupropion hydroxylation in a concentration-dependent manner. Ethanol 24-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 17709370-5 2007 Acetonitrile, DMSO, and ethanol were shown to increase the Km and decrease the intrinsic clearance (CLint) of CYP2B6-mediated bupropion hydroxylation in a concentration-dependent manner. Bupropion 126-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 17709370-14 2007 Methanol and acetonitrile at concentrations of < or =0.5% and < or =1% (v/v) appeared to be suitable for the measurement of CYP2B6- and CYP2C8-mediated activities, respectively. Methanol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 17709370-14 2007 Methanol and acetonitrile at concentrations of < or =0.5% and < or =1% (v/v) appeared to be suitable for the measurement of CYP2B6- and CYP2C8-mediated activities, respectively. acetonitrile 13-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 18090046-1 2007 BACKGROUND: Cytochrome P450 2B6 (CYP2B6)-G516T genotype is associated with altered activity of hepatic CYP2B6 and efavirenz pharmacokinetics, but the relationship between the CYP2B6-G516T genotype and nevirapine (NVP) pharmacokinetics in plasma and cerebrospinal fluid (CSF) is limited. Nevirapine 201-211 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-31 18090046-1 2007 BACKGROUND: Cytochrome P450 2B6 (CYP2B6)-G516T genotype is associated with altered activity of hepatic CYP2B6 and efavirenz pharmacokinetics, but the relationship between the CYP2B6-G516T genotype and nevirapine (NVP) pharmacokinetics in plasma and cerebrospinal fluid (CSF) is limited. Nevirapine 201-211 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 18090046-1 2007 BACKGROUND: Cytochrome P450 2B6 (CYP2B6)-G516T genotype is associated with altered activity of hepatic CYP2B6 and efavirenz pharmacokinetics, but the relationship between the CYP2B6-G516T genotype and nevirapine (NVP) pharmacokinetics in plasma and cerebrospinal fluid (CSF) is limited. Nevirapine 201-211 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 18090046-1 2007 BACKGROUND: Cytochrome P450 2B6 (CYP2B6)-G516T genotype is associated with altered activity of hepatic CYP2B6 and efavirenz pharmacokinetics, but the relationship between the CYP2B6-G516T genotype and nevirapine (NVP) pharmacokinetics in plasma and cerebrospinal fluid (CSF) is limited. Nevirapine 201-211 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 17693640-2 2007 At a cyt P450:cyt b(5) molar ratio of 1:1 under single turnover conditions, cyt P450 2B4 catalyzes the oxidation of the substrates, benzphetamine and cyclohexane, with rate constants of 18 +/- 2 and 29 +/- 4.5 s(-1), respectively. Benzphetamine 132-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-13 17693640-2 2007 At a cyt P450:cyt b(5) molar ratio of 1:1 under single turnover conditions, cyt P450 2B4 catalyzes the oxidation of the substrates, benzphetamine and cyclohexane, with rate constants of 18 +/- 2 and 29 +/- 4.5 s(-1), respectively. Benzphetamine 132-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 17693640-2 2007 At a cyt P450:cyt b(5) molar ratio of 1:1 under single turnover conditions, cyt P450 2B4 catalyzes the oxidation of the substrates, benzphetamine and cyclohexane, with rate constants of 18 +/- 2 and 29 +/- 4.5 s(-1), respectively. Cyclohexane 150-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-13 17693640-2 2007 At a cyt P450:cyt b(5) molar ratio of 1:1 under single turnover conditions, cyt P450 2B4 catalyzes the oxidation of the substrates, benzphetamine and cyclohexane, with rate constants of 18 +/- 2 and 29 +/- 4.5 s(-1), respectively. Cyclohexane 150-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 17693640-4 2007 In marked contrast, at a cyt P450:CPR molar ratio of 1:1, cyt P450 2B4 catalyzes the oxidation of benzphetamine congruent with100-fold (k = 0.15 +/- 0.05 s(-1)) and cyclohexane congruent with10-fold (k = 2.5 +/- 0.35 s(-1)) more slowly. Benzphetamine 98-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 17693640-4 2007 In marked contrast, at a cyt P450:CPR molar ratio of 1:1, cyt P450 2B4 catalyzes the oxidation of benzphetamine congruent with100-fold (k = 0.15 +/- 0.05 s(-1)) and cyclohexane congruent with10-fold (k = 2.5 +/- 0.35 s(-1)) more slowly. Benzphetamine 98-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 17693640-4 2007 In marked contrast, at a cyt P450:CPR molar ratio of 1:1, cyt P450 2B4 catalyzes the oxidation of benzphetamine congruent with100-fold (k = 0.15 +/- 0.05 s(-1)) and cyclohexane congruent with10-fold (k = 2.5 +/- 0.35 s(-1)) more slowly. Cyclohexane 165-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 17693640-4 2007 In marked contrast, at a cyt P450:CPR molar ratio of 1:1, cyt P450 2B4 catalyzes the oxidation of benzphetamine congruent with100-fold (k = 0.15 +/- 0.05 s(-1)) and cyclohexane congruent with10-fold (k = 2.5 +/- 0.35 s(-1)) more slowly. Cyclohexane 165-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 17693640-8 2007 Under steady-state conditions at all cyt b(5):cyt P450 molar ratios examined, cyt b(5) inhibits the rate of NADPH consumption. NADP 108-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 17693640-9 2007 Nevertheless, at low cyt b(5):cyt P450 molar ratios <or=1:1, the rate of metabolism of cyclohexane and benzphetamine is enhanced, whereas at higher cyt b(5):cyt P450 molar ratios, cyt b(5) progressively inhibits both NADPH consumption and the rate of metabolism. Cyclohexane 90-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 17693640-9 2007 Nevertheless, at low cyt b(5):cyt P450 molar ratios <or=1:1, the rate of metabolism of cyclohexane and benzphetamine is enhanced, whereas at higher cyt b(5):cyt P450 molar ratios, cyt b(5) progressively inhibits both NADPH consumption and the rate of metabolism. Cyclohexane 90-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-168 17693640-9 2007 Nevertheless, at low cyt b(5):cyt P450 molar ratios <or=1:1, the rate of metabolism of cyclohexane and benzphetamine is enhanced, whereas at higher cyt b(5):cyt P450 molar ratios, cyt b(5) progressively inhibits both NADPH consumption and the rate of metabolism. Benzphetamine 106-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 17693640-9 2007 Nevertheless, at low cyt b(5):cyt P450 molar ratios <or=1:1, the rate of metabolism of cyclohexane and benzphetamine is enhanced, whereas at higher cyt b(5):cyt P450 molar ratios, cyt b(5) progressively inhibits both NADPH consumption and the rate of metabolism. Benzphetamine 106-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-168 17907788-0 2007 Peroxynitrite inactivation of human cytochrome P450s 2B6 and 2E1: heme modification and site-specific nitrotyrosine formation. Peroxynitrous Acid 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-64 17907788-0 2007 Peroxynitrite inactivation of human cytochrome P450s 2B6 and 2E1: heme modification and site-specific nitrotyrosine formation. Heme 66-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-64 17907788-0 2007 Peroxynitrite inactivation of human cytochrome P450s 2B6 and 2E1: heme modification and site-specific nitrotyrosine formation. 3-nitrotyrosine 102-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-64 17715394-0 2007 Rational engineering of human cytochrome P450 2B6 for enhanced expression and stability: importance of a Leu264->Phe substitution. Phenylalanine 116-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-49 17256129-11 2007 DPIC, an inhibitor of P450 Red, had no effect on cell growth inhibition by RH1 in T47D cells, and had only a small effect on cell growth inhibition by RH1 in the presence of the NQO1 inhibitor, dicoumarol, in T47D-P450 cells. dpic 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 17979764-2 2007 P450 is a membrane-anchored protein that shows a variety of interaction with membrane phospholipids, which affect the membrane topology and catalytic activities of the protein. Phospholipids 86-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 17979764-3 2007 In particular, anionic phospholipids, nonbilayer forming lipids, and the degree of saturation of the lipid fatty acyl chain play important roles in the functional regulation of P450, as well as in the bilayer structure of the membrane. Phospholipids 23-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-181 17979764-4 2007 However, despite the importance of phospholipids in the regulation of P450s, the interaction of the protein with membrane phospholipids, and the membrane properties induced by phospholipids which regulate P450, are unclear. Phospholipids 35-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 17979764-5 2007 In this review, we describe the effect of the physicochemical properties of the phospholipid constituents of biological membranes on hepatic P450 catalytic activity, membrane insertion (and/or penetration), and structural changes. Phospholipids 80-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 17223085-0 2007 CYP2B6 genotype alters abstinence rates in a bupropion smoking cessation trial. Bupropion 45-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 17223085-9 2007 CONCLUSIONS: These data suggest that smokers with the CYP2B6*6 genotype have a higher liability to relapse on placebo and that they may be good candidates for bupropion treatment for smoking cessation. Bupropion 159-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 17223085-1 2007 BACKGROUND: CYP2B6 is the primary enzyme involved in bupropion (Zyban; GlaxoSmithKline, Research Triangle Park, North Carolina) metabolism. Bupropion 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 17223085-1 2007 BACKGROUND: CYP2B6 is the primary enzyme involved in bupropion (Zyban; GlaxoSmithKline, Research Triangle Park, North Carolina) metabolism. Bupropion 64-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 17223085-2 2007 Genetic polymorphisms in CYP2B6, such as CYP2B6*6, can alter bupropion metabolism and may affect bupropion treatment outcome. Bupropion 61-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 17223085-2 2007 Genetic polymorphisms in CYP2B6, such as CYP2B6*6, can alter bupropion metabolism and may affect bupropion treatment outcome. Bupropion 61-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 17223085-2 2007 Genetic polymorphisms in CYP2B6, such as CYP2B6*6, can alter bupropion metabolism and may affect bupropion treatment outcome. Bupropion 97-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 17223085-2 2007 Genetic polymorphisms in CYP2B6, such as CYP2B6*6, can alter bupropion metabolism and may affect bupropion treatment outcome. Bupropion 97-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 17576809-0 2007 Identification of rat and human cytochrome p450 isoforms and a rat serum esterase that metabolize the pyrethroid insecticides deltamethrin and esfenvalerate. Pyrethrins 102-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 17344806-2 2007 Analysis of liver microsomes from patients treated with metamizole revealed selectively higher expression of cytochromes P450, CYP2B6 and CYP3A4 (3.8- and 2.8-fold, respectively), and 2.9-fold higher bupropion hydroxylase activity compared with untreated subjects. Dipyrone 56-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 17576809-0 2007 Identification of rat and human cytochrome p450 isoforms and a rat serum esterase that metabolize the pyrethroid insecticides deltamethrin and esfenvalerate. decamethrin 126-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 17591676-3 2007 DHEA treatment elevated the expression and activities of CYP3A4, CYP2C9, CYP2C19, and CYP2B6 in primary cultures of human hepatocytes. Dehydroepiandrosterone 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 17591676-7 2007 Because CYP2B6 induction by DHEA in human hepatocytes might involve either PXR or constitutive androstane receptor (CAR) activation, we performed experiments in primary hepatocytes from CAR knockout mice and observed that CAR was required for maximal induction of Cyp2b10 by DHEA. Dehydroepiandrosterone 28-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 17598095-2 2007 METHODS: Methadone was incubated with various drugs (n = 10) and cDNA-expressed CYP3A4, CYP2D6, CYP2B6, CYP2C19 and CYP1A2 enzymes to screen for their inhibition potency. Methadone 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 17576809-0 2007 Identification of rat and human cytochrome p450 isoforms and a rat serum esterase that metabolize the pyrethroid insecticides deltamethrin and esfenvalerate. fenvalerate 143-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 17591676-10 2007 Elucidation of CAR activation and subsequent induction of CYP2B6 by DHEA presented an additional mechanism by which the sterol can modify the expression of P450s. Dehydroepiandrosterone 68-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 17591676-10 2007 Elucidation of CAR activation and subsequent induction of CYP2B6 by DHEA presented an additional mechanism by which the sterol can modify the expression of P450s. Sterols 120-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 17576809-3 2007 Using a parent depletion approach, rat and human cytochromes P450 (P450s) were screened for their ability to eliminate deltamethrin or esfenvalerate during in vitro incubations. decamethrin 119-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 17576809-3 2007 Using a parent depletion approach, rat and human cytochromes P450 (P450s) were screened for their ability to eliminate deltamethrin or esfenvalerate during in vitro incubations. decamethrin 119-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-72 17576809-3 2007 Using a parent depletion approach, rat and human cytochromes P450 (P450s) were screened for their ability to eliminate deltamethrin or esfenvalerate during in vitro incubations. fenvalerate 135-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 17576809-3 2007 Using a parent depletion approach, rat and human cytochromes P450 (P450s) were screened for their ability to eliminate deltamethrin or esfenvalerate during in vitro incubations. fenvalerate 135-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-72 17576809-4 2007 Rat P450 isoforms CYP1A1, CYP2C6, CYP2C11, and CYP3A2 and human P450 isoforms CYP2C8, CYP2C19, and CYP3A5 were capable of metabolizing either pyrethroid. Pyrethrins 142-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-8 17576809-4 2007 Rat P450 isoforms CYP1A1, CYP2C6, CYP2C11, and CYP3A2 and human P450 isoforms CYP2C8, CYP2C19, and CYP3A5 were capable of metabolizing either pyrethroid. Pyrethrins 142-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 17576809-6 2007 Rat and human P450s that metabolize esfenvalerate and deltamethrin do so with similar kinetics. fenvalerate 36-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-19 17576809-6 2007 Rat and human P450s that metabolize esfenvalerate and deltamethrin do so with similar kinetics. decamethrin 54-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-19 17576809-11 2007 These studies suggest that the difference in rates of oxidative metabolism of pyrethroids by rat and human hepatic microsomes is dependent on the expression levels of individual P450 isoforms rather than their specific activity. Pyrethrins 78-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 17591676-0 2007 Dehydroepiandrosterone induces human CYP2B6 through the constitutive androstane receptor. Dehydroepiandrosterone 0-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 17992728-1 2007 The metabolism of (+)-fenchol was investigated in vitro using liver microsomes of rats and humans and recombinant cytochrome P450 (P450 or CYP) enzymes in insect cells in which human/rat P450 and NADPH-P450 reductase cDNAs had been introduced. fenchol 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-142 17992728-3 2007 (+)-Fenchol was oxidized to fenchone by human liver microsomal P450 enzymes. fenchone 28-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 17992728-1 2007 The metabolism of (+)-fenchol was investigated in vitro using liver microsomes of rats and humans and recombinant cytochrome P450 (P450 or CYP) enzymes in insect cells in which human/rat P450 and NADPH-P450 reductase cDNAs had been introduced. fenchol 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-129 17992728-1 2007 The metabolism of (+)-fenchol was investigated in vitro using liver microsomes of rats and humans and recombinant cytochrome P450 (P450 or CYP) enzymes in insect cells in which human/rat P450 and NADPH-P450 reductase cDNAs had been introduced. fenchol 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-135 17992728-9 2007 Human recombinant CYP2A6-catalyzed (+)-fenchol oxidation with a Vmax value of 6.96 nmol min-1 nmol-1 P450 and apparent Km value of 0.09 mM. fenchol 35-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 17992728-3 2007 (+)-Fenchol was oxidized to fenchone by human liver microsomal P450 enzymes. fenchol 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 17992728-13 2007 Kinetic analysis showed that the Km values for the formation of fenchone, 6-exo- hydroxyfenchol and 10-hydroxyfenchol in rats treated with phenobarbital were 0.06, 0.03 and 0.03 mM, and Vmax values were 2.94, 6.1 and 13.8 nmol min-1 nmol-1 P450, respectively. fenchone 64-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-244 17672514-0 2007 In vitro bioactivation of 3-(N-phenylamino)propane-1,2-diol by human and rat liver microsomes and recombinant P450 enzymes. 3-(n-phenylamino)propane-1,2-diol 26-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-114 17992728-13 2007 Kinetic analysis showed that the Km values for the formation of fenchone, 6-exo- hydroxyfenchol and 10-hydroxyfenchol in rats treated with phenobarbital were 0.06, 0.03 and 0.03 mM, and Vmax values were 2.94, 6.1 and 13.8 nmol min-1 nmol-1 P450, respectively. 6-exo- hydroxyfenchol 74-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-244 17992728-13 2007 Kinetic analysis showed that the Km values for the formation of fenchone, 6-exo- hydroxyfenchol and 10-hydroxyfenchol in rats treated with phenobarbital were 0.06, 0.03 and 0.03 mM, and Vmax values were 2.94, 6.1 and 13.8 nmol min-1 nmol-1 P450, respectively. 10-hydroxyfenchol 100-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-244 17992728-13 2007 Kinetic analysis showed that the Km values for the formation of fenchone, 6-exo- hydroxyfenchol and 10-hydroxyfenchol in rats treated with phenobarbital were 0.06, 0.03 and 0.03 mM, and Vmax values were 2.94, 6.1 and 13.8 nmol min-1 nmol-1 P450, respectively. Phenobarbital 139-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-244 17335546-1 2007 AIMS: To investigate the effect of kidney disease on bupropion pharmacokinetics and on cytochrome P450 (CYP) 2B6 activity as measured by bupropion hydroxylation. Bupropion 137-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-112 17686225-0 2007 Efavirenz-induced neurological symptoms in rare homozygote CYP2B6 *2/*2 (C64T). efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 17686225-3 2007 For example, CYP2B6 *6 (G516T and A785G) and *7 (G516T, A785G and C1459T) prolonged the EFV half-life despite discontinuation of EFV. efavirenz 88-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 17356468-1 2007 BACKGROUND: The CYP2B6-G516T polymorphism has been shown to alter plasma efavirenz (EFV) concentrations in adults. efavirenz 84-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 17431031-0 2007 Cytochrome P450 eicosanoids are activators of peroxisome proliferator-activated receptor alpha. Eicosanoids 16-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 17431031-1 2007 Cytochrome P450 (P450) eicosanoids regulate vascular tone, renal tubular transport, cellular proliferation, and inflammation. Eicosanoids 23-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 17431031-4 2007 We show here that P450 eicosanoids can bind to and activate PPARalpha and result in the modulation of PPARalpha target gene expression. Eicosanoids 23-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-22 17431031-8 2007 In addition, P450 eicosanoids induced CYP4A1, sEH, and CYP2C11 expression, suggesting that they can regulate their own levels. Eicosanoids 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 17431031-9 2007 Given that P450 eicosanoids have multiple cardiovascular effects, pharmacological modulation of their formation and/or degradation may yield therapeutic benefits. Eicosanoids 16-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 17620218-0 2007 Involvement of human cytochrome P450 2B6 in the omega- and 4-hydroxylation of the anesthetic agent propofol. Propofol 99-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-40 17638512-2 2007 One of the less well-studied human cytochrome P450s is (CYP)2B6, a homologue of the rodent phenobarbital-inducible CYP2B enzymes. Phenobarbital 91-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-63 17638512-2 2007 One of the less well-studied human cytochrome P450s is (CYP)2B6, a homologue of the rodent phenobarbital-inducible CYP2B enzymes. Phenobarbital 91-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-120 17620218-2 2007 Of six cDNA-expressed human P450 enzymes tested, CYP2B6 and CYP1A2, followed by CYP3A4, had high catalytic activities at a 20 microM propofol concentration, corresponding to clinical plasma levels. Propofol 133-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 17620218-7 2007 These results suggest that CYP2B6 has an important role in propofol omega- and 4-hydroxylation in human livers and that the hepatic contents of CYP2B6, CYP3A4, and CYP1A2 determine which P450 enzymes play major roles in propofol oxidation in individual humans. Propofol 220-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 17620218-3 2007 K(m) and k(cat) values for propofol omega- and 4-hydroxyation were 27 microM and 21 nmol omega-hydroxypropofol formed/min/nmol CYP2B6 and 30 microM and 42 nmol 4-hydroxypropofol formed/min/nmol CYP2B6, respectively. Propofol 27-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 17620218-7 2007 These results suggest that CYP2B6 has an important role in propofol omega- and 4-hydroxylation in human livers and that the hepatic contents of CYP2B6, CYP3A4, and CYP1A2 determine which P450 enzymes play major roles in propofol oxidation in individual humans. Propofol 220-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-150 17620218-3 2007 K(m) and k(cat) values for propofol omega- and 4-hydroxyation were 27 microM and 21 nmol omega-hydroxypropofol formed/min/nmol CYP2B6 and 30 microM and 42 nmol 4-hydroxypropofol formed/min/nmol CYP2B6, respectively. Propofol 27-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 194-200 17620218-3 2007 K(m) and k(cat) values for propofol omega- and 4-hydroxyation were 27 microM and 21 nmol omega-hydroxypropofol formed/min/nmol CYP2B6 and 30 microM and 42 nmol 4-hydroxypropofol formed/min/nmol CYP2B6, respectively. omega-hydroxypropofol 89-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 17620218-3 2007 K(m) and k(cat) values for propofol omega- and 4-hydroxyation were 27 microM and 21 nmol omega-hydroxypropofol formed/min/nmol CYP2B6 and 30 microM and 42 nmol 4-hydroxypropofol formed/min/nmol CYP2B6, respectively. omega-hydroxypropofol 89-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 194-200 17620218-4 2007 CYP2B6 expressed in HepG2 cells also effectively catalyzed propofol omega- and 4-hydroxylation. Propofol 59-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 17620218-5 2007 In a panel of individual human liver microsomes, propofol omega- and 4-hydroxylation activities (at the substrate concentration of 20 microM) were highly correlated with CYP2B6 contents, and moderately with CYP3A4 contents. Propofol 49-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-176 17455229-2 2007 Among them, Cytochrome P450 2B6 (CYP2B6) plays a critical role in the deactivation of testosterone. Testosterone 86-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-31 17620218-7 2007 These results suggest that CYP2B6 has an important role in propofol omega- and 4-hydroxylation in human livers and that the hepatic contents of CYP2B6, CYP3A4, and CYP1A2 determine which P450 enzymes play major roles in propofol oxidation in individual humans. Propofol 59-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 17455229-2 2007 Among them, Cytochrome P450 2B6 (CYP2B6) plays a critical role in the deactivation of testosterone. Testosterone 86-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 17584015-1 2007 Cytochrome P450 constitute a superfamily of heme-containing enzymes that catalyze the oxidative biotransformation of structurally diverse xenobiotics including drugs. Heme 44-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 17455229-11 2007 Furthermore, overexpression of CYP2B6 in LNCaP cells significantly decreased testosterone-induced proliferation. Testosterone 77-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 17433262-1 2007 Involvement of cytochrome P450 (P450 or CYP) 2C19, 2C9, and 3A4 in N-oxidation of voriconazole, a new triazole antifungal agent, has been demonstrated using human liver microsomes. Voriconazole 82-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 17433262-1 2007 Involvement of cytochrome P450 (P450 or CYP) 2C19, 2C9, and 3A4 in N-oxidation of voriconazole, a new triazole antifungal agent, has been demonstrated using human liver microsomes. Voriconazole 82-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-49 17433262-1 2007 Involvement of cytochrome P450 (P450 or CYP) 2C19, 2C9, and 3A4 in N-oxidation of voriconazole, a new triazole antifungal agent, has been demonstrated using human liver microsomes. Triazoles 102-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 17433262-1 2007 Involvement of cytochrome P450 (P450 or CYP) 2C19, 2C9, and 3A4 in N-oxidation of voriconazole, a new triazole antifungal agent, has been demonstrated using human liver microsomes. Triazoles 102-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-49 17433262-3 2007 Among recombinant P450 isoforms using Escherichia coli expression systems, CYP2C19 and CYP3A4 had voriconazole N-oxidation activities, but not CYP2C9. Voriconazole 98-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-22 17433521-4 2007 Curcumin inhibited CYP1A2 (IC(50), 40.0 microM), CYP3A4 (IC(50), 16.3 microM), CYP2D6 (IC(50), 50.3 microM), CYP2C9 (IC(50), 4.3 microM) and CYP2B6 (IC(50), 24.5 microM). Curcumin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 17433521-5 2007 Curcumin showed a competitive type of inhibition towards CYP1A2, CYP3A4 and CYP2B6, whereas a non-competitive type of inhibition was observed with respect to CYP2D6 and CYP2C9. Curcumin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 17559344-4 2007 The formation rate of 8-hydroxyefavirenz correlated significantly with CYP2B6 protein (Spearman"s r(S) = 0.54; p < 0.0001) and bupropion hydroxylase activity (r(S) = 0.73; p < 0.0001). 8-hydroxyefavirenz 22-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 17559344-8 2007 We found no effect of gender and age on any of the phenotypes tested, but prior exposure to carbamazepine markedly increased CYP2B6 protein expression and activity as well as efavirenz 8-hydroxylation. Carbamazepine 92-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 17502835-0 2007 Genetic polymorphisms of CYP2B6 affect the pharmacokinetics/pharmacodynamics of cyclophosphamide in Japanese cancer patients. Cyclophosphamide 80-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 17502835-7 2007 We found that the homozygotes of CYP2B6*6 (Q172H and K262R) showed significantly (P<0.05) higher clearance and shorter half-life of cyclophosphamide than heterozygotes and homozygotes of CYP2B6*1. Cyclophosphamide 135-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 17502835-12 2007 CONCLUSIONS: We clarified that the single nucleotide polymorphisms in the promoter region or introns in the CYP2B6 affect the potency of cyclophosphamide activation to 4-hydroxycyclophosphamide. Cyclophosphamide 137-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 17502835-12 2007 CONCLUSIONS: We clarified that the single nucleotide polymorphisms in the promoter region or introns in the CYP2B6 affect the potency of cyclophosphamide activation to 4-hydroxycyclophosphamide. 4-hydroxycyclophosphamide 168-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 17584015-4 2007 Mechanism-based P450 inactivation usually involves bioactivation of the xenobiotic to a reactive intermediate, which covalently modifies an active site amino acid residue and/or coordinates to the heme prosthetic group. Heme 197-201 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 17361128-3 2007 Clopidogrel"s activation involves two sequential steps by CYP3A, CYP1A2, CYP2C9, CYP2C19, and/or CYP2B6. Clopidogrel 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 17329992-4 2007 As CYP2B6 slow metabolizer (SM) status results in a reduced ability to metabolize (S)-methadone, electrocardiograms, CYP2B6 genotypes, and (R)- and (S)-methadone plasma concentrations were obtained for 179 patients receiving (R,S)-methadone. Methadone 82-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 3-9 17329992-4 2007 As CYP2B6 slow metabolizer (SM) status results in a reduced ability to metabolize (S)-methadone, electrocardiograms, CYP2B6 genotypes, and (R)- and (S)-methadone plasma concentrations were obtained for 179 patients receiving (R,S)-methadone. Methadone 148-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 3-9 17329992-4 2007 As CYP2B6 slow metabolizer (SM) status results in a reduced ability to metabolize (S)-methadone, electrocardiograms, CYP2B6 genotypes, and (R)- and (S)-methadone plasma concentrations were obtained for 179 patients receiving (R,S)-methadone. (r,s)-methadone 225-240 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 3-9 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 2 subfamily B member 6 Homo sapiens 11-15 17325652-6 2007 KEY RESULTS: Three compounds proved to be very potent and selective competitive inhibitors of CYP2B6 in vitro (IC(50)<1 microM): 4-(4-chlorobenzyl)pyridine (CBP), 4-(4-nitrobenzyl)pyridine (NBP), and 4-benzylpyridine (BP). 4-(4-Chlorobenzyl)pyridine 132-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 17376532-1 2007 The present study demonstrates direct electron transfer between cytochromes P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51b1) on the one hand and screen-printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB) on the other. Graphite 186-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 17376532-1 2007 The present study demonstrates direct electron transfer between cytochromes P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51b1) on the one hand and screen-printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB) on the other. Graphite 186-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 17376532-1 2007 The present study demonstrates direct electron transfer between cytochromes P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51b1) on the one hand and screen-printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB) on the other. didodecyldimethylammonium 244-277 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 17376532-1 2007 The present study demonstrates direct electron transfer between cytochromes P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51b1) on the one hand and screen-printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB) on the other. didodecyldimethylammonium 244-277 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 17376532-1 2007 The present study demonstrates direct electron transfer between cytochromes P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51b1) on the one hand and screen-printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB) on the other. didodecyldimethylammonium 279-283 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 17376532-1 2007 The present study demonstrates direct electron transfer between cytochromes P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51b1) on the one hand and screen-printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB) on the other. didodecyldimethylammonium 279-283 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 17376532-3 2007 Electron transfer, direct electrochemical reduction and interaction with P450 substrates (oxygen, benzphetamine, and lanosterol) and with P450 inhibitor (ketoconazole) were analyzed using cyclic voltammetry (CV), square wave voltammetry (SWV) differential pulse voltammetry (DPV), and amperometry. Ketoconazole 154-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-142 17325652-6 2007 KEY RESULTS: Three compounds proved to be very potent and selective competitive inhibitors of CYP2B6 in vitro (IC(50)<1 microM): 4-(4-chlorobenzyl)pyridine (CBP), 4-(4-nitrobenzyl)pyridine (NBP), and 4-benzylpyridine (BP). 4-(4-nitrobenzyl)pyridine 166-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 17325652-6 2007 KEY RESULTS: Three compounds proved to be very potent and selective competitive inhibitors of CYP2B6 in vitro (IC(50)<1 microM): 4-(4-chlorobenzyl)pyridine (CBP), 4-(4-nitrobenzyl)pyridine (NBP), and 4-benzylpyridine (BP). 4-(4-nitrobenzyl)pyridine 193-196 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 17325652-6 2007 KEY RESULTS: Three compounds proved to be very potent and selective competitive inhibitors of CYP2B6 in vitro (IC(50)<1 microM): 4-(4-chlorobenzyl)pyridine (CBP), 4-(4-nitrobenzyl)pyridine (NBP), and 4-benzylpyridine (BP). 4,4'-Bis(N-carbazolyl)-1,1'-biphenyl 160-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 17325652-6 2007 KEY RESULTS: Three compounds proved to be very potent and selective competitive inhibitors of CYP2B6 in vitro (IC(50)<1 microM): 4-(4-chlorobenzyl)pyridine (CBP), 4-(4-nitrobenzyl)pyridine (NBP), and 4-benzylpyridine (BP). 4-benzylpyridine 203-219 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 17325652-6 2007 KEY RESULTS: Three compounds proved to be very potent and selective competitive inhibitors of CYP2B6 in vitro (IC(50)<1 microM): 4-(4-chlorobenzyl)pyridine (CBP), 4-(4-nitrobenzyl)pyridine (NBP), and 4-benzylpyridine (BP). 4-benzylpyridine 161-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 17235330-1 2007 To assess the association of CYP2B6 allelic diversity with efavirenz (EFV) pharmacokinetics, we performed extensive genotyping of 15 relevant single nucleotide polymorphism in 169 study participants, and full resequencing of CYP2B6 in individuals with abnormal EFV plasma levels. efavirenz 59-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 17235330-1 2007 To assess the association of CYP2B6 allelic diversity with efavirenz (EFV) pharmacokinetics, we performed extensive genotyping of 15 relevant single nucleotide polymorphism in 169 study participants, and full resequencing of CYP2B6 in individuals with abnormal EFV plasma levels. efavirenz 70-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 17495414-1 2007 Although there is evidence in the literature of the participation of CYP2B6 in the metabolism of selegiline, it is not clear which other CYP isoforms contribute to its metabolism. Selegiline 97-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 17495414-6 2007 These findings confirm that CYP2B6 plays a major role in the metabolism of selegiline and also suggest the involvement of CYP3A4 and CYP2A6. Selegiline 75-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 17259447-3 2007 N-Demethylation of methadone in vitro is predominantly mediated by cytochrome P450 CYP3A4 and CYP2B6 and somewhat by CYP2C19. Nitrogen 0-1 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 17259447-3 2007 N-Demethylation of methadone in vitro is predominantly mediated by cytochrome P450 CYP3A4 and CYP2B6 and somewhat by CYP2C19. Methadone 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 17259447-4 2007 This investigation evaluated stereoselectivity, models, and kinetic parameters for methadone N-demethylation by recombinant CYP2B6, CYP3A4, and CYP2C19, and the potential for interactions between enantiomers during racemate metabolism. Methadone 83-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-130 17259447-4 2007 This investigation evaluated stereoselectivity, models, and kinetic parameters for methadone N-demethylation by recombinant CYP2B6, CYP3A4, and CYP2C19, and the potential for interactions between enantiomers during racemate metabolism. Nitrogen 93-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-130 17259447-9 2007 For CYP2B6, the interaction between enantiomers was stereoselective, with S-methadone as a more potent inhibitor of R-methadone N-demethylation than R-of S-methadone. Methadone 74-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 17259447-9 2007 For CYP2B6, the interaction between enantiomers was stereoselective, with S-methadone as a more potent inhibitor of R-methadone N-demethylation than R-of S-methadone. Methadone 116-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 17259447-9 2007 For CYP2B6, the interaction between enantiomers was stereoselective, with S-methadone as a more potent inhibitor of R-methadone N-demethylation than R-of S-methadone. Methadone 154-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 17259447-13 2007 Changes in plasma R/S methadone ratios observed after rifampin or troleandomycin pretreatment in humans in vivo were successfully predicted by CYP2B6- but not CYP3A4-catalyzed methadone N-demethylation. Methadone 22-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 17259447-13 2007 Changes in plasma R/S methadone ratios observed after rifampin or troleandomycin pretreatment in humans in vivo were successfully predicted by CYP2B6- but not CYP3A4-catalyzed methadone N-demethylation. Rifampin 54-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 17259447-13 2007 Changes in plasma R/S methadone ratios observed after rifampin or troleandomycin pretreatment in humans in vivo were successfully predicted by CYP2B6- but not CYP3A4-catalyzed methadone N-demethylation. Troleandomycin 66-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 17259447-14 2007 CYP2B6 is a predominant catalyst of stereoselective methadone metabolism in vitro. Methadone 52-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 17259447-15 2007 In vivo, CYP2B6 may be a major determinant of methadone metabolism and disposition, and CYP2B6 activity and stereoselective metabolic interactions may confer variability in methadone disposition. Methadone 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 17259447-15 2007 In vivo, CYP2B6 may be a major determinant of methadone metabolism and disposition, and CYP2B6 activity and stereoselective metabolic interactions may confer variability in methadone disposition. Methadone 173-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 17292855-2 2007 Three PAH were found to be substrates for the oxidation by P450(BSbeta), namely anthracene, 9-methyl-anthracene and azulene. Polycyclic Aromatic Hydrocarbons 6-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-71 17292855-2 2007 Three PAH were found to be substrates for the oxidation by P450(BSbeta), namely anthracene, 9-methyl-anthracene and azulene. anthracene 80-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-71 17292855-2 2007 Three PAH were found to be substrates for the oxidation by P450(BSbeta), namely anthracene, 9-methyl-anthracene and azulene. 9-methylanthracene 92-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-71 17292855-2 2007 Three PAH were found to be substrates for the oxidation by P450(BSbeta), namely anthracene, 9-methyl-anthracene and azulene. azulene 116-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-71 17292855-5 2007 The stability of P450(BSbeta) against hydrogen peroxide, cumene, and ter-butyl hydroperoxide was determined. Hydrogen Peroxide 38-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-29 17292855-5 2007 The stability of P450(BSbeta) against hydrogen peroxide, cumene, and ter-butyl hydroperoxide was determined. cumene 57-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-29 17292855-5 2007 The stability of P450(BSbeta) against hydrogen peroxide, cumene, and ter-butyl hydroperoxide was determined. ter-butyl hydroperoxide 69-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-29 17213193-2 2007 Here, we describe the detailed characterization of a functionally homogeneous 1:1 complex of cytochrome P450 3A4 (CYP3A4) and cytochrome P450 reductase solubilized via self-assembly in a nanoscale phospholipid bilayer. Phospholipids 197-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-108 17213193-7 2007 The resolution of the fractional contributions of binding intermediates of CYP3A4 into experimentally observed overall spin shift and the rates of steady-state NADPH oxidation and product formation provide new detailed insight into the mechanisms of cooperativity and allosteric regulation in this human cytochrome P450. NADP 160-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 315-319 17123739-0 2007 Structural analysis of cytochromes P450 shows differences in flexibility of heme 2- and 4-vinyls. Heme 76-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 17123739-1 2007 Structural analysis of the orientations of heme vinyl side chains was carried out using the published crystallographic data for different cytochromes P450. Heme 43-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 17123739-8 2007 Differences between P450 forms suggest a variability in heme-region flexibility and in communication with the rest of enzyme. Heme 56-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 21591074-6 2007 Specifically, the activity of human P450 1A2, which converts caffeine into paraxanthine, can be investigated by measuring the change in caffeine and paraxanthine concentrations in urine over time following a single dose of caffeine. Caffeine 61-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 21591074-6 2007 Specifically, the activity of human P450 1A2, which converts caffeine into paraxanthine, can be investigated by measuring the change in caffeine and paraxanthine concentrations in urine over time following a single dose of caffeine. 1,7-dimethylxanthine 75-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 21591074-6 2007 Specifically, the activity of human P450 1A2, which converts caffeine into paraxanthine, can be investigated by measuring the change in caffeine and paraxanthine concentrations in urine over time following a single dose of caffeine. Caffeine 136-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 21591074-6 2007 Specifically, the activity of human P450 1A2, which converts caffeine into paraxanthine, can be investigated by measuring the change in caffeine and paraxanthine concentrations in urine over time following a single dose of caffeine. 1,7-dimethylxanthine 149-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 21591074-6 2007 Specifically, the activity of human P450 1A2, which converts caffeine into paraxanthine, can be investigated by measuring the change in caffeine and paraxanthine concentrations in urine over time following a single dose of caffeine. Caffeine 136-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 17142561-4 2007 Bupropion hydroxylation, amodiaquine N-deethylation, and midazolam 1"-hydroxylation were chosen as probe reactions for CYP2B6, CYP2C8, and CYP3A5 and were analyzed using liquid chromatography-tandem mass spectrometry. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 17352764-11 2007 CONCLUSIONS: CYP2B6 G516T significantly influenced nevirapine trough concentrations in HIV-infected patients in Uganda. Nevirapine 51-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 17268056-3 2007 (-)-Camphor was oxidized to 5-exo-hydroxyfenchone by human liver microsomal cytochrome (P450) enzymes. Camphor 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-92 17160483-8 2007 CYP2B6, CYP2C19, and CYP2D6 all contributed to the formation of N-methylPEA, while only CYP2B6 catalyzed the formation of N-propargylPEA. n-propargylpea 122-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 17160483-8 2007 CYP2B6, CYP2C19, and CYP2D6 all contributed to the formation of N-methylPEA, while only CYP2B6 catalyzed the formation of N-propargylPEA. n-propargylpea 122-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 17079358-0 2007 Human hepatic cytochrome p450-specific metabolism of parathion and chlorpyrifos. Parathion 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 17079358-0 2007 Human hepatic cytochrome p450-specific metabolism of parathion and chlorpyrifos. Chlorpyrifos 67-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 17079358-2 2007 Thiophosphorus OPs, once bioactivated by cytochromes P450 (P450s), form oxon metabolites, which are potent acetylcholinesterase inhibitors. thiophosphorus ops 0-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 17079358-2 2007 Thiophosphorus OPs, once bioactivated by cytochromes P450 (P450s), form oxon metabolites, which are potent acetylcholinesterase inhibitors. thiophosphorus ops 0-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-64 17079358-2 2007 Thiophosphorus OPs, once bioactivated by cytochromes P450 (P450s), form oxon metabolites, which are potent acetylcholinesterase inhibitors. oxon 72-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 17079358-2 2007 Thiophosphorus OPs, once bioactivated by cytochromes P450 (P450s), form oxon metabolites, which are potent acetylcholinesterase inhibitors. oxon 72-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-64 17079358-7 2007 With K(m) and V(max) values of 0.61 microM, 4827 pmol/min/nmol P450 and 0.81 microM, 12,544 pmol/min/nmol for formation of paraoxon and chlorpyrifos-oxon, respectively, CYP2B6 favored the desulfation reaction. Paraoxon 123-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 17079358-7 2007 With K(m) and V(max) values of 0.61 microM, 4827 pmol/min/nmol P450 and 0.81 microM, 12,544 pmol/min/nmol for formation of paraoxon and chlorpyrifos-oxon, respectively, CYP2B6 favored the desulfation reaction. Paraoxon 123-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 17079358-7 2007 With K(m) and V(max) values of 0.61 microM, 4827 pmol/min/nmol P450 and 0.81 microM, 12,544 pmol/min/nmol for formation of paraoxon and chlorpyrifos-oxon, respectively, CYP2B6 favored the desulfation reaction. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 136-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 17079358-7 2007 With K(m) and V(max) values of 0.61 microM, 4827 pmol/min/nmol P450 and 0.81 microM, 12,544 pmol/min/nmol for formation of paraoxon and chlorpyrifos-oxon, respectively, CYP2B6 favored the desulfation reaction. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 136-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 17045554-7 2007 Several studies on efavirenz, a commonly used antiretroviral drug, have reported higher plasma exposure and early side effects with the homozygous variant of the hepatic cytochrome P450 enzyme CYP2B6 G516T polymorphism, which are more frequently found in African-American subjects. efavirenz 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-199 17270273-10 2007 The implications of inhibition of P450-catalzyed oxidation steps that are known or speculated to influence arachadonic acid, cholesterol, and catecholamine neurotransmitters pathways in human brain will be considered. arachadonic acid 107-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 17270273-10 2007 The implications of inhibition of P450-catalzyed oxidation steps that are known or speculated to influence arachadonic acid, cholesterol, and catecholamine neurotransmitters pathways in human brain will be considered. Cholesterol 125-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 17270273-10 2007 The implications of inhibition of P450-catalzyed oxidation steps that are known or speculated to influence arachadonic acid, cholesterol, and catecholamine neurotransmitters pathways in human brain will be considered. Catecholamines 142-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 17268056-3 2007 (-)-Camphor was oxidized to 5-exo-hydroxyfenchone by human liver microsomal cytochrome (P450) enzymes. 5-exo-hydroxyfenchone 28-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-92 17268056-6 2007 First, of eleven recombinant human P450 enzymes tested, CYP2A6 catalyzed the oxidation of (-)-camphor. Camphor 90-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 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 2 subfamily B member 6 Homo sapiens 12-18 17484521-0 2007 Metabolism of (-)-fenchone by CYP2A6 and CYP2B6 in human liver microsomes. fenchone 14-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 17484521-5 2007 CYP2A6 and CYP2B6 were major enzymes involved in the hydroxylation of (-)-fenchone by human liver microsomes, based on the following lines of evidence. fenchone 70-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 17484521-6 2007 First, of 11 recombinant human P450 enzymes tested, CYP2A6 and CYP2B6 catalysed the oxidation of (-)-fenchone. fenchone 97-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 17484521-8 2007 Finally, there was a good correlation between CYP2A6, CYP2B6 contents and (-)-fenchone hydroxylation activities in liver microsomes of 11 human samples. fenchone 74-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 17503755-5 2007 However, under conditions of limited EFV metabolism, that is, the group of 23 individuals carrying two copies of CYP2B6 loss/diminished-function alleles, plasma AUC values were highest among individuals receiving TFV (n=5, 353,031 ng*h/ml), compared with those not receiving TFV (n=18, 180,689 ng*h/ml). Tenofovir 213-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 17503755-5 2007 However, under conditions of limited EFV metabolism, that is, the group of 23 individuals carrying two copies of CYP2B6 loss/diminished-function alleles, plasma AUC values were highest among individuals receiving TFV (n=5, 353,031 ng*h/ml), compared with those not receiving TFV (n=18, 180,689 ng*h/ml). Tenofovir 275-278 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 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 2 subfamily B member 6 Homo sapiens 12-18 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 2 subfamily B member 6 Homo sapiens 12-18 18447001-6 2007 CYP2B6, the most efficient isoform for BALC production, was inhibited by CPO, while CYP3A4, the most efficient isoform for ET production, was activated by CPO. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 73-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 18447001-6 2007 CYP2B6, the most efficient isoform for BALC production, was inhibited by CPO, while CYP3A4, the most efficient isoform for ET production, was activated by CPO. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 155-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 18447001-7 2007 CPO inhibited CYP2B6 production of both BALC and ET from DEET, but activated CYP3A4 production of ET, while inhibiting CYP3A4 BALC production. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 18447001-7 2007 CPO inhibited CYP2B6 production of both BALC and ET from DEET, but activated CYP3A4 production of ET, while inhibiting CYP3A4 BALC production. N-ethyl-3-toluamide 49-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 21783735-2 2007 The mean inhibitory concentrations (IC(50)) for 7-pentoxyresorufin-O-dealkylation (CYP2B) and bupropion hydroxylation (2B6) were 48.9 and 41.7muM, respectively. 7-pentoxyresorufin-o 48-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-88 17050648-0 2007 CYP2B6, CYP2D6, and CYP3A4 catalyze the primary oxidative metabolism of perhexiline enantiomers by human liver microsomes. Perhexiline 72-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 17020957-2 2007 However, ketoconazole at therapeutic, high concentrations also inhibits cytochromes P450 (P450) other than CYP3A4/5, which has made the predictions of DDIs less accurate. Ketoconazole 9-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 17020957-2 2007 However, ketoconazole at therapeutic, high concentrations also inhibits cytochromes P450 (P450) other than CYP3A4/5, which has made the predictions of DDIs less accurate. Ketoconazole 9-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-94 17020957-11 2007 Thus, this approach provides a simple method to more precisely predict the DDIs for P450 substrates when coadministered with ketoconazole or any other competitive P450 inhibitors in humans. Ketoconazole 125-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 17115150-11 2007 It is important to determine if these differences, along with previously reported differences in cytochrome P450 2B6 allele frequencies, are associated with altered metabolism/effectiveness of artemisinin drugs. artemisinin 193-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-116 21783735-8 2007 Among the nine major human CYPs, CYP3A4 was the only one responsible for metalaxyl hydroxylation, while CYP2B6 was the major isoform responsible for (di)demethylation and lactone formation. Lactones 171-178 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-110 17073778-0 2006 Protein dynamics and imidazole binding in cytochrome P450 enzymes. imidazole 21-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 17427179-5 2007 CPS significantly inhibited the metabolism of fipronil by CYP3A4 as well as the metabolism of nonane by CYP2B6. Chlorpyrifos 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-110 17427179-5 2007 CPS significantly inhibited the metabolism of fipronil by CYP3A4 as well as the metabolism of nonane by CYP2B6. nonane 94-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-110 17084830-4 2006 CYP2B6 and 3A5 were also induced by fipronil, although at lower levels (2-3-fold). fipronil 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 17062778-3 2007 This study describes the identification of human cytochrome P450 (P450) and UDP-glucuronosyltransferase (UGT) enzymes involved in the in vitro metabolism of muraglitazar. muraglitazar 157-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-71 17164128-1 2007 The three major enzyme systems, cyclo-oxygenase, lipoxygenase, and cytochrome P450 (P450/CYP), metabolize arachidonic acid (AA) to biologically active compounds. Arachidonic Acid 106-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 17164128-1 2007 The three major enzyme systems, cyclo-oxygenase, lipoxygenase, and cytochrome P450 (P450/CYP), metabolize arachidonic acid (AA) to biologically active compounds. Arachidonic Acid 106-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-92 17164128-3 2007 The four regioisomeric eicosanoids, 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs) of AA metabolites derived by P450 epoxygenases have shown to possess potent biological effects in numerous tissues. Eicosanoids 23-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 17164128-3 2007 The four regioisomeric eicosanoids, 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids (EETs) of AA metabolites derived by P450 epoxygenases have shown to possess potent biological effects in numerous tissues. 5,6-, 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids 36-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 17073778-1 2006 P450 (cytochrome P450) enzymes have major roles in the biosynthesis of endogenous factors such as steroids and eicosanoids, in the termination of the action of endogenous factors such as retinoic acid, in the metabolism of most drugs and xenobiotics and in the generation of toxic and carcinogenic products. Steroids 98-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 17073778-1 2006 P450 (cytochrome P450) enzymes have major roles in the biosynthesis of endogenous factors such as steroids and eicosanoids, in the termination of the action of endogenous factors such as retinoic acid, in the metabolism of most drugs and xenobiotics and in the generation of toxic and carcinogenic products. Steroids 98-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 17073778-1 2006 P450 (cytochrome P450) enzymes have major roles in the biosynthesis of endogenous factors such as steroids and eicosanoids, in the termination of the action of endogenous factors such as retinoic acid, in the metabolism of most drugs and xenobiotics and in the generation of toxic and carcinogenic products. Eicosanoids 111-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 17073778-1 2006 P450 (cytochrome P450) enzymes have major roles in the biosynthesis of endogenous factors such as steroids and eicosanoids, in the termination of the action of endogenous factors such as retinoic acid, in the metabolism of most drugs and xenobiotics and in the generation of toxic and carcinogenic products. Eicosanoids 111-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 17073778-1 2006 P450 (cytochrome P450) enzymes have major roles in the biosynthesis of endogenous factors such as steroids and eicosanoids, in the termination of the action of endogenous factors such as retinoic acid, in the metabolism of most drugs and xenobiotics and in the generation of toxic and carcinogenic products. Tretinoin 187-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 17073778-1 2006 P450 (cytochrome P450) enzymes have major roles in the biosynthesis of endogenous factors such as steroids and eicosanoids, in the termination of the action of endogenous factors such as retinoic acid, in the metabolism of most drugs and xenobiotics and in the generation of toxic and carcinogenic products. Tretinoin 187-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 17073778-3 2006 The crystallographic analysis of the deformability of two bacterial P450 active sites associated with the binding of azole (a class of inhibitors with an imidazole or triazole ring that co-ordinates to the haem iron) inhibitors described in the present study illustrates the importance of protein conformational malleability in the binding of imidazole derivatives. Azoles 117-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 17142962-0 2006 Metabolism of (+)-fenchone by CYP2A6 and CYP2B6 in human liver microsomes. fenchone 14-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 17073778-3 2006 The crystallographic analysis of the deformability of two bacterial P450 active sites associated with the binding of azole (a class of inhibitors with an imidazole or triazole ring that co-ordinates to the haem iron) inhibitors described in the present study illustrates the importance of protein conformational malleability in the binding of imidazole derivatives. imidazole 154-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 17142962-5 2006 CYP2A6 and CYP2B6 in human liver microsomes were major enzymes involved in the hydroxylation of (+)-fenchone, based on the following lines of evidence. fenchone 96-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 17142962-6 2006 First, of eleven recombinant human P450 enzymes tested, CYP2A6 and CYP2B6 catalyzed oxidation of (+)-fenchone. fenchone 97-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 17073778-3 2006 The crystallographic analysis of the deformability of two bacterial P450 active sites associated with the binding of azole (a class of inhibitors with an imidazole or triazole ring that co-ordinates to the haem iron) inhibitors described in the present study illustrates the importance of protein conformational malleability in the binding of imidazole derivatives. Triazoles 167-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 17142962-7 2006 Second, oxidation of (+)-fenchone was inhibited by thioTEPA, (+)-menthofuran anti-CYP2A6 and anti-CYP2B6 antibodies. fenchone 21-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 17073778-3 2006 The crystallographic analysis of the deformability of two bacterial P450 active sites associated with the binding of azole (a class of inhibitors with an imidazole or triazole ring that co-ordinates to the haem iron) inhibitors described in the present study illustrates the importance of protein conformational malleability in the binding of imidazole derivatives. Iron 211-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 17142962-8 2006 Finally, there was a good correlation between CYP2A6, CYP2B6 contents and (+)-fenchone hydroxylation activities in liver microsomes of 8 human samples. fenchone 74-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 17073778-3 2006 The crystallographic analysis of the deformability of two bacterial P450 active sites associated with the binding of azole (a class of inhibitors with an imidazole or triazole ring that co-ordinates to the haem iron) inhibitors described in the present study illustrates the importance of protein conformational malleability in the binding of imidazole derivatives. imidazole 343-352 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 17073780-2 2006 These include the first characterized sterol 14alpha-demethylase P450 (CYP51), a known target for azole and triazole drugs in yeasts and fungi. Azoles 98-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 17073780-2 2006 These include the first characterized sterol 14alpha-demethylase P450 (CYP51), a known target for azole and triazole drugs in yeasts and fungi. Triazoles 108-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 17073780-4 2006 The CYP121 P450 shows structural relationships with P450 enzymes involved in synthesis of polyketide antibiotics. polyketide antibiotics 90-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 17073780-4 2006 The CYP121 P450 shows structural relationships with P450 enzymes involved in synthesis of polyketide antibiotics. polyketide antibiotics 90-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 17073780-7 2006 This paper reviews our current knowledge of these and the other P450 systems in Mtb including recent data relating to the reversible conversion of the CYP51 enzyme between P450 (thiolate-co-ordinated) and P420 (thiol-co-ordinated) species on reduction of the haem iron in the absence of a P450 substrate. thiolate 178-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 17073780-7 2006 This paper reviews our current knowledge of these and the other P450 systems in Mtb including recent data relating to the reversible conversion of the CYP51 enzyme between P450 (thiolate-co-ordinated) and P420 (thiol-co-ordinated) species on reduction of the haem iron in the absence of a P450 substrate. thiolate 178-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 17073780-7 2006 This paper reviews our current knowledge of these and the other P450 systems in Mtb including recent data relating to the reversible conversion of the CYP51 enzyme between P450 (thiolate-co-ordinated) and P420 (thiol-co-ordinated) species on reduction of the haem iron in the absence of a P450 substrate. thiolate 178-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 17073780-7 2006 This paper reviews our current knowledge of these and the other P450 systems in Mtb including recent data relating to the reversible conversion of the CYP51 enzyme between P450 (thiolate-co-ordinated) and P420 (thiol-co-ordinated) species on reduction of the haem iron in the absence of a P450 substrate. Sulfhydryl Compounds 178-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 17073780-7 2006 This paper reviews our current knowledge of these and the other P450 systems in Mtb including recent data relating to the reversible conversion of the CYP51 enzyme between P450 (thiolate-co-ordinated) and P420 (thiol-co-ordinated) species on reduction of the haem iron in the absence of a P450 substrate. Sulfhydryl Compounds 178-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 17073780-7 2006 This paper reviews our current knowledge of these and the other P450 systems in Mtb including recent data relating to the reversible conversion of the CYP51 enzyme between P450 (thiolate-co-ordinated) and P420 (thiol-co-ordinated) species on reduction of the haem iron in the absence of a P450 substrate. Sulfhydryl Compounds 178-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 17073780-7 2006 This paper reviews our current knowledge of these and the other P450 systems in Mtb including recent data relating to the reversible conversion of the CYP51 enzyme between P450 (thiolate-co-ordinated) and P420 (thiol-co-ordinated) species on reduction of the haem iron in the absence of a P450 substrate. Iron 264-268 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 17073780-7 2006 This paper reviews our current knowledge of these and the other P450 systems in Mtb including recent data relating to the reversible conversion of the CYP51 enzyme between P450 (thiolate-co-ordinated) and P420 (thiol-co-ordinated) species on reduction of the haem iron in the absence of a P450 substrate. Iron 264-268 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 17073780-7 2006 This paper reviews our current knowledge of these and the other P450 systems in Mtb including recent data relating to the reversible conversion of the CYP51 enzyme between P450 (thiolate-co-ordinated) and P420 (thiol-co-ordinated) species on reduction of the haem iron in the absence of a P450 substrate. Iron 264-268 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 17073780-8 2006 The accessory flavoprotein and iron-sulfur proteins required to drive P450 catalysis are also discussed, providing an overview of the current state of knowledge of Mtb P450 redox systems. Iron 31-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 17073780-8 2006 The accessory flavoprotein and iron-sulfur proteins required to drive P450 catalysis are also discussed, providing an overview of the current state of knowledge of Mtb P450 redox systems. Sulfur 36-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 17178267-6 2006 As previously reported, CYP2B6*6/*6 carriers had significantly higher trough (S)-methadone plasma levels (P = .0001) and a trend toward higher (R)-methadone plasma levels (P = .07). Methadone 77-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 17178267-6 2006 As previously reported, CYP2B6*6/*6 carriers had significantly higher trough (S)-methadone plasma levels (P = .0001) and a trend toward higher (R)-methadone plasma levels (P = .07). Methadone 143-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 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 2 subfamily B member 6 Homo sapiens 32-38 16872679-1 2006 By participating in pathways of cholesterol biosynthesis and elimination, different cytochrome P450 (P450 or CYP) enzymes play an important role in maintenance of cholesterol homeostasis. Cholesterol 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-112 16956955-0 2006 Identification of binding sites of non-I-helix water molecules in mammalian cytochromes p450. Water 47-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 16956955-2 2006 At the molecular level, the details of P450 catalysis are still under investigation, but the importance of water-mediated proton shuttles seems evident in the catalytic cycle as it progresses through various heme iron-oxygen enzyme intermediates. Oxygen 218-224 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 16956955-3 2006 The study of P450-bound waters has been largely restricted to bacterial enzymes that may or may not reflect the location or function of waters in human drug-metabolizing P450s. Water 24-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 16956955-3 2006 The study of P450-bound waters has been largely restricted to bacterial enzymes that may or may not reflect the location or function of waters in human drug-metabolizing P450s. Water 136-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-175 16956955-5 2006 Described herein is the identification of seven well defined water clusters in mammalian P450s identified by calculating the density of globally aligned waters as reported by Tanner and coworkers [Bottoms CA, White TA, and Tanner JJ (2006) Proteins 64:404-421 (DOI: 10.1002/prot.21014)]. Water 61-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-94 16956955-6 2006 All water binding sites were in or within the immediate vicinity of the active sites of the P450s, but most were not near the conserved I-helix threonine often implicated in P450 catalysis. Water 4-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-97 16956955-6 2006 All water binding sites were in or within the immediate vicinity of the active sites of the P450s, but most were not near the conserved I-helix threonine often implicated in P450 catalysis. Water 4-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 16956955-7 2006 Therefore, it is possible that some of the water binding sites identified here ultimately determine P450 catalytic efficiency either by working as an extension of the I-helix water network, or by acting in novel proton shuttles that modulate the nonproductive shunting of reactive oxygen species. Water 43-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-104 16956955-7 2006 Therefore, it is possible that some of the water binding sites identified here ultimately determine P450 catalytic efficiency either by working as an extension of the I-helix water network, or by acting in novel proton shuttles that modulate the nonproductive shunting of reactive oxygen species. Water 175-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-104 16956955-7 2006 Therefore, it is possible that some of the water binding sites identified here ultimately determine P450 catalytic efficiency either by working as an extension of the I-helix water network, or by acting in novel proton shuttles that modulate the nonproductive shunting of reactive oxygen species. Reactive Oxygen Species 272-295 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-104 16988011-1 2006 Our previous studies have suggested a role for AMP-activated protein kinase (AMPK) in the induction of CYP2B6 by phenobarbital (PB) in hepatoma-derived cells (Rencurel et al., 2005). Phenobarbital 113-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 16988011-1 2006 Our previous studies have suggested a role for AMP-activated protein kinase (AMPK) in the induction of CYP2B6 by phenobarbital (PB) in hepatoma-derived cells (Rencurel et al., 2005). Phenobarbital 128-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 16988011-2 2006 In this study, we showed in primary human hepatocytes that: 1) 5"-phosphoribosyl-5-aminoimidazol-4-carboxamide 1-beta-d-ribofuranoside and the biguanide metformin, known activators of AMPK, dose-dependently increase the expression of CYP2B6 and CYP3A4 to an extent similar to that of PB. 1) 5"-phosphoribosyl-5-aminoimidazol-4-carboxamide 1-beta-d-ribofuranoside 60-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 234-240 16988011-2 2006 In this study, we showed in primary human hepatocytes that: 1) 5"-phosphoribosyl-5-aminoimidazol-4-carboxamide 1-beta-d-ribofuranoside and the biguanide metformin, known activators of AMPK, dose-dependently increase the expression of CYP2B6 and CYP3A4 to an extent similar to that of PB. Biguanides 143-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 234-240 16997911-0 2006 Structure-activity relationship and elucidation of the determinant factor(s) responsible for the mechanism-based inactivation of cytochrome P450 2B6 by substituted phenyl diaziridines. phenyl diaziridines 164-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-148 16997911-4 2006 In this report, the structure-activity relationships for the mechanism-based inactivation of cytochrome P450 2B6 by a series of aryl diaziridines were investigated. aryl diaziridines 128-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-112 17101742-1 2006 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of drugs such as bupropion, efavirenz, propofol, and selegiline, among others. Bupropion 76-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 17101742-1 2006 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of drugs such as bupropion, efavirenz, propofol, and selegiline, among others. Bupropion 76-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 17101742-1 2006 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of drugs such as bupropion, efavirenz, propofol, and selegiline, among others. efavirenz 87-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 17101742-1 2006 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of drugs such as bupropion, efavirenz, propofol, and selegiline, among others. efavirenz 87-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 17101742-1 2006 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of drugs such as bupropion, efavirenz, propofol, and selegiline, among others. Propofol 98-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 17101742-1 2006 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of drugs such as bupropion, efavirenz, propofol, and selegiline, among others. Propofol 98-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 17101742-1 2006 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of drugs such as bupropion, efavirenz, propofol, and selegiline, among others. Selegiline 112-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 17101742-1 2006 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of drugs such as bupropion, efavirenz, propofol, and selegiline, among others. Selegiline 112-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 16872679-1 2006 By participating in pathways of cholesterol biosynthesis and elimination, different cytochrome P450 (P450 or CYP) enzymes play an important role in maintenance of cholesterol homeostasis. Cholesterol 163-174 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-112 16945988-0 2006 Contribution of CYP2C9, CYP2A6, and CYP2B6 to valproic acid metabolism in hepatic microsomes from individuals with the CYP2C9*1/*1 genotype. Valproic Acid 46-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 16945988-2 2006 cDNA-expressed CYP2C9*1, CYP2A6, and CYP2B6 were the most active catalysts of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA formation. 2-propyl-4-pentenoic acid 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 16945988-2 2006 cDNA-expressed CYP2C9*1, CYP2A6, and CYP2B6 were the most active catalysts of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA formation. 4-Hydroxyvalproic acid 89-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 16945988-2 2006 cDNA-expressed CYP2C9*1, CYP2A6, and CYP2B6 were the most active catalysts of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA formation. 5-Hydroxyvalproic acid 103-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 16945988-4 2006 CYP2A6 was the most active in catalyzing VPA 3-hydroxylation, whereas CYP1A1, CYP2B6, CYP4F2, and CYP4F3B were less active. Valproic Acid 41-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 17047492-10 2006 Among 155 participants who received efavirenz without nelfinavir, virologic failure was associated with a two-locus interaction between ABCB1 2677G>T and CYP2B6 516G>T (65% accuracy, P<0.001). efavirenz 36-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 17164692-7 2006 Although roxithromycin did not favor the generation of toxic metabolites from cyclophosphamide, it led to cyclophosphamide accumulation due to inhibition of both CYP3A4 and CYP2B6. Roxithromycin 9-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 17119049-9 2006 Differentially expressed probe sets within the nitrosamine metabolism pathway were consistently underexpressed (100%), with strong effects observed for NQ01, CYP2B6, and CYP2E1. Nitrosamines 47-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-164 16882766-0 2006 Synthesis of substituted phenyl diaziridines and characterization as mechanism-based inactivators of human cytochrome P450 2B6. phenyl diaziridines 25-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-126 16882766-1 2006 The metabolism of arylhydrazines by cytochromes P450 (P450s) has previously been shown to yield aryl-iron complexes that inhibit P450 enzymes as a result of heme modification. arylhydrazines 18-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 16882766-1 2006 The metabolism of arylhydrazines by cytochromes P450 (P450s) has previously been shown to yield aryl-iron complexes that inhibit P450 enzymes as a result of heme modification. arylhydrazines 18-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-59 16882766-1 2006 The metabolism of arylhydrazines by cytochromes P450 (P450s) has previously been shown to yield aryl-iron complexes that inhibit P450 enzymes as a result of heme modification. arylhydrazines 18-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 16882766-1 2006 The metabolism of arylhydrazines by cytochromes P450 (P450s) has previously been shown to yield aryl-iron complexes that inhibit P450 enzymes as a result of heme modification. aryl-iron 96-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 16882766-1 2006 The metabolism of arylhydrazines by cytochromes P450 (P450s) has previously been shown to yield aryl-iron complexes that inhibit P450 enzymes as a result of heme modification. aryl-iron 96-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-59 16882766-1 2006 The metabolism of arylhydrazines by cytochromes P450 (P450s) has previously been shown to yield aryl-iron complexes that inhibit P450 enzymes as a result of heme modification. aryl-iron 96-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 16882766-1 2006 The metabolism of arylhydrazines by cytochromes P450 (P450s) has previously been shown to yield aryl-iron complexes that inhibit P450 enzymes as a result of heme modification. Heme 157-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 16882766-1 2006 The metabolism of arylhydrazines by cytochromes P450 (P450s) has previously been shown to yield aryl-iron complexes that inhibit P450 enzymes as a result of heme modification. Heme 157-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-59 16882766-1 2006 The metabolism of arylhydrazines by cytochromes P450 (P450s) has previously been shown to yield aryl-iron complexes that inhibit P450 enzymes as a result of heme modification. Heme 157-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 16882766-2 2006 These modifications of the heme have been used to probe the topology of the active site of several P450s. Heme 27-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-104 16882766-3 2006 Therefore, diaziridines containing one or more substitutions on the phenyl ring were synthesized and evaluated as potential mechanism-based inactivators of P450 2B enzymes that could be used to elucidate the active site topology. diaziridines 11-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 156-160 16882766-4 2006 Five of the six trifluoroaryldiaziridines tested selectively inactivated P450 2B6 in the reconstituted system in a time-, concentration-, and NADPH-dependent manner as measured using the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation assay. trifluoroaryldiaziridines 16-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 16882766-4 2006 Five of the six trifluoroaryldiaziridines tested selectively inactivated P450 2B6 in the reconstituted system in a time-, concentration-, and NADPH-dependent manner as measured using the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation assay. NADP 142-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 16882766-4 2006 Five of the six trifluoroaryldiaziridines tested selectively inactivated P450 2B6 in the reconstituted system in a time-, concentration-, and NADPH-dependent manner as measured using the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation assay. 7-ethoxy-4-trifluoromethylcoumarin 187-223 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 16882766-6 2006 Analysis of the P450 heme from P450s inactivated by the five substituted diaziridines suggested that the activity loss was not due to heme destruction as measured by the reduced-CO spectrum or high-performance liquid chromatography of the P450 heme. Heme 21-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 16882766-6 2006 Analysis of the P450 heme from P450s inactivated by the five substituted diaziridines suggested that the activity loss was not due to heme destruction as measured by the reduced-CO spectrum or high-performance liquid chromatography of the P450 heme. Heme 21-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-36 16882766-6 2006 Analysis of the P450 heme from P450s inactivated by the five substituted diaziridines suggested that the activity loss was not due to heme destruction as measured by the reduced-CO spectrum or high-performance liquid chromatography of the P450 heme. Heme 21-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 16882766-6 2006 Analysis of the P450 heme from P450s inactivated by the five substituted diaziridines suggested that the activity loss was not due to heme destruction as measured by the reduced-CO spectrum or high-performance liquid chromatography of the P450 heme. diaziridines 73-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 16882766-6 2006 Analysis of the P450 heme from P450s inactivated by the five substituted diaziridines suggested that the activity loss was not due to heme destruction as measured by the reduced-CO spectrum or high-performance liquid chromatography of the P450 heme. diaziridines 73-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-36 16882766-6 2006 Analysis of the P450 heme from P450s inactivated by the five substituted diaziridines suggested that the activity loss was not due to heme destruction as measured by the reduced-CO spectrum or high-performance liquid chromatography of the P450 heme. diaziridines 73-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 16882766-7 2006 Dialysis experiments indicated the irreversible nature of the inactivation and the reaction between the diaziridine compounds and the P450 enzyme. Diaziridine 104-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-138 16882766-8 2006 Interestingly, a thiomethyl-substituted phenyl diaziridine had no effect on the activity of P450 2B6 in the reconstituted system, but competitively inhibited the O-debenzylation activity of P450 3A4 with 7-benzyloxy-4-(trifluoromethyl)coumarin as substrate. thiomethyl-substituted phenyl diaziridine 17-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 190-194 16882766-8 2006 Interestingly, a thiomethyl-substituted phenyl diaziridine had no effect on the activity of P450 2B6 in the reconstituted system, but competitively inhibited the O-debenzylation activity of P450 3A4 with 7-benzyloxy-4-(trifluoromethyl)coumarin as substrate. 7-benzyloxy-4-trifluoromethylcoumarin 204-243 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 190-194 16882766-9 2006 Binding spectra suggest that this compound bound reversibly to P450 2B6, and preliminary results indicate that 3-(4-methylthiophenyl)-3-(trifluoromethyl)diaziridine is metabolized by P450 2B6. 3-(4-methylthiophenyl)-3-(trifluoromethyl)diaziridine 111-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 16882766-9 2006 Binding spectra suggest that this compound bound reversibly to P450 2B6, and preliminary results indicate that 3-(4-methylthiophenyl)-3-(trifluoromethyl)diaziridine is metabolized by P450 2B6. 3-(4-methylthiophenyl)-3-(trifluoromethyl)diaziridine 111-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 16943206-3 2006 Furthermore, the crystal structures of imidazole-bound P450(cam) and the 1-methylimidazole-bound P450(cam) L244A mutant reveal that the ligands have distinct binding modes in the two proteins. imidazole 39-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 16943206-3 2006 Furthermore, the crystal structures of imidazole-bound P450(cam) and the 1-methylimidazole-bound P450(cam) L244A mutant reveal that the ligands have distinct binding modes in the two proteins. 1-methylimidazole 73-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 16959210-0 2006 Cysteine 98 in CYP3A4 contributes to conformational integrity required for P450 interaction with CYP reductase. Cysteine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 16959210-14 2006 In addition, the C98W mutant exhibited a 41% decrease in the maximum electron flow rate between P450 and reductase as measured by reduced nicotinamide adenine dinucleotide phosphate consumption at a saturating reductase concentration. NADP 138-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-114 16959210-15 2006 In conclusion, our data strongly suggest that cysteine 98 in the B-C loop region significantly contributes to conformational integrity and catalytic activity of CYP3A4, and that this residue or residues nearby might be involved in an interaction with P450 reductase. Cysteine 46-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 251-255 16763015-7 2006 Hepatocytes exposed to 1 mM FFA for 14 h showed lower activity values of CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2D6, CYP2E1, and CYP3A4 enzymes than nontreated hepatocytes (about 45-65% reduction). Fatty Acids, Nonesterified 28-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-95 17073575-1 2006 CYP2B6 metabolizes a number of drug substrates, that are usually non-planar, neutral or weakly basic, fairly lipophilic with one or two hydrogen bond acceptors, on which it catalyses various oxidative reactions. Hydrogen 136-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 17073575-2 2006 For bupropion, cyclophosphamide, ifosfamide, pethidine, ketamine and propofol, these reactions represent major metabolic or activation pathways and for their kinetics CYP2B6 function is of considerable importance. Bupropion 4-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-173 17073575-2 2006 For bupropion, cyclophosphamide, ifosfamide, pethidine, ketamine and propofol, these reactions represent major metabolic or activation pathways and for their kinetics CYP2B6 function is of considerable importance. Cyclophosphamide 15-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-173 17073575-2 2006 For bupropion, cyclophosphamide, ifosfamide, pethidine, ketamine and propofol, these reactions represent major metabolic or activation pathways and for their kinetics CYP2B6 function is of considerable importance. Ifosfamide 33-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-173 17073575-2 2006 For bupropion, cyclophosphamide, ifosfamide, pethidine, ketamine and propofol, these reactions represent major metabolic or activation pathways and for their kinetics CYP2B6 function is of considerable importance. Meperidine 45-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-173 17073575-2 2006 For bupropion, cyclophosphamide, ifosfamide, pethidine, ketamine and propofol, these reactions represent major metabolic or activation pathways and for their kinetics CYP2B6 function is of considerable importance. Ketamine 56-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-173 17073575-2 2006 For bupropion, cyclophosphamide, ifosfamide, pethidine, ketamine and propofol, these reactions represent major metabolic or activation pathways and for their kinetics CYP2B6 function is of considerable importance. Propofol 69-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-173 17073575-5 2006 Thiotepa is the most selective of the inhibitors, but is not useful as an in vivo inhibitor, whereas ticlopidine and clopidogrel can be used as CYP2B6-selective probes in human clinical studies. Clopidogrel 117-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-150 17073575-6 2006 Bupropion hydroxylation is a selective, and consequently useful, in vivo probe for CYP2B6. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 17073575-8 2006 With validated in vitro and in vivo substrates (e.g. bupropion) and inhibitors (e.g. ticlopidine), it is expected that pharmacological (including pharmacogenetic) and clinical significance of CYP2B6 will be delineated more fully in the near future. Bupropion 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 17073575-8 2006 With validated in vitro and in vivo substrates (e.g. bupropion) and inhibitors (e.g. ticlopidine), it is expected that pharmacological (including pharmacogenetic) and clinical significance of CYP2B6 will be delineated more fully in the near future. Ticlopidine 85-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-198 17180920-1 2006 The present study demonstrates the direct electron transfer between cytochrome P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51MT) and screen printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB). Graphite 173-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 17180920-1 2006 The present study demonstrates the direct electron transfer between cytochrome P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51MT) and screen printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB). Graphite 173-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 17180920-1 2006 The present study demonstrates the direct electron transfer between cytochrome P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51MT) and screen printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB). didodecyldimethylammonium 231-264 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 17180920-1 2006 The present study demonstrates the direct electron transfer between cytochrome P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51MT) and screen printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB). didodecyldimethylammonium 231-264 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 17180920-1 2006 The present study demonstrates the direct electron transfer between cytochrome P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51MT) and screen printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB). didodecyldimethylammonium 266-270 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 17180920-1 2006 The present study demonstrates the direct electron transfer between cytochrome P450 2B4 (CYP2B4), P450 1A2 (CYP1A2), sterol 14alpha-demethylase (CYP51MT) and screen printed graphite electrodes, modified with gold nanoparticles and didodecyldimethylammonium bromide (DDAB). didodecyldimethylammonium 266-270 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 17180920-2 2006 Electrodetection of heme proteins is possible when 2-200 pmol P450/electrode were adsorbed on the surface of nanostructured electrochemical interfaces. Heme 20-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 16855053-3 2006 Using 16 individual human liver microsomes (HLMs), a strong correlation was observed with endosulfan sulfate formation and S-mephenytoin N-demethylase activity of CYP2B6 (r(2) = 0.79), whereas a moderate correlation with testosterone 6 beta-hydroxylase activity of CYP3A4 (r(2) = 0.54) was observed. endosulfan sulfate 90-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 16855053-4 2006 Ticlopidine (5 microM), a potent CYP2B6 inhibitor, and ketoconazole (10 microM), a selective CYP3A4 inhibitor, together inhibited approximately 90% of endosulfan-alpha metabolism in HLMs. Ticlopidine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 16855053-6 2006 In five of the six HLMs used, the percentage inhibition with ticlopidine and ketoconazole in the same incubation correlated with the combined % TNRs for CYP2B6 and CYP3A4. Ticlopidine 61-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 16855053-6 2006 In five of the six HLMs used, the percentage inhibition with ticlopidine and ketoconazole in the same incubation correlated with the combined % TNRs for CYP2B6 and CYP3A4. Ketoconazole 77-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 16855053-7 2006 This study shows that endosulfan-alpha is metabolized by HLMs to a single metabolite, endosulfan sulfate, and that it has potential use, in combination with inhibitors, as an in vitro probe for CYP2B6 and 3A4 catalytic activities. Endosulfan 22-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 194-200 17216026-1 2006 The cytochrome P450 enzymes (P450s or CYPs) form a large family of heme proteins involved in drug metabolism and in the biosynthesis of steroids, lipids, vitamins and natural products. Heme 67-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 17216026-1 2006 The cytochrome P450 enzymes (P450s or CYPs) form a large family of heme proteins involved in drug metabolism and in the biosynthesis of steroids, lipids, vitamins and natural products. Steroids 136-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 16782764-0 2006 Mechanism of inactivation of human cytochrome P450 2B6 by phencyclidine. Phencyclidine 58-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-54 16918308-4 2006 Potential mechanisms for oxazaphosphorine resistance include decreased activation by cytochrome P450s (e.g. CYP3A4, CYP2C9 and CYP2B6), increased deactivation of the agents by deactivating enzymes such as aldehyde dehydrogenases (ALDHs), increased cellular thiol level, increased DNA repair capacity, and altered cellular apoptotic response to DNA repair, e.g. deficient apoptosis due to lack of cellular mechanisms to result in cell death following DNA damage. oxazaphosphorine 25-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 17009913-2 2006 Bupropion undergoes metabolic transformation to an active metabolite, 4-hydroxybupropion, through hepatic cytochrome P450-2B6 (CYP2B6) and has inhibitory effects on cytochrome P450-2D6 (CYP2D6), thus raising concern for clinically-relevant drug interactions. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-125 17009913-2 2006 Bupropion undergoes metabolic transformation to an active metabolite, 4-hydroxybupropion, through hepatic cytochrome P450-2B6 (CYP2B6) and has inhibitory effects on cytochrome P450-2D6 (CYP2D6), thus raising concern for clinically-relevant drug interactions. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 17009913-2 2006 Bupropion undergoes metabolic transformation to an active metabolite, 4-hydroxybupropion, through hepatic cytochrome P450-2B6 (CYP2B6) and has inhibitory effects on cytochrome P450-2D6 (CYP2D6), thus raising concern for clinically-relevant drug interactions. hydroxybupropion 70-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-125 17009913-2 2006 Bupropion undergoes metabolic transformation to an active metabolite, 4-hydroxybupropion, through hepatic cytochrome P450-2B6 (CYP2B6) and has inhibitory effects on cytochrome P450-2D6 (CYP2D6), thus raising concern for clinically-relevant drug interactions. hydroxybupropion 70-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 17003847-3 2006 Since nevirapine-induced hepatotoxicity commonly occurs between 2-12 weeks of treatment, and nevirapine is a known inducer of human CYP3A and CYP2B6 isozymes, it was envisaged that the hepatotoxicity was due to activation of nevirapine to toxic metabolites by the induced enzymes. Nevirapine 93-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 17003847-3 2006 Since nevirapine-induced hepatotoxicity commonly occurs between 2-12 weeks of treatment, and nevirapine is a known inducer of human CYP3A and CYP2B6 isozymes, it was envisaged that the hepatotoxicity was due to activation of nevirapine to toxic metabolites by the induced enzymes. Nevirapine 93-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 16623664-1 2006 CAR (constitutive active/androstane receptor) regulates both the distal enhancer PBREM (phenobarbital-responsive enhancer module) and the proximal element OARE [OA (okadaic acid) response element] to synergistically up-regulate the endogenous CYP2B6 (where CYP is cytochrome P450) gene in HepG2 cells. Phenobarbital 88-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 243-249 16623664-1 2006 CAR (constitutive active/androstane receptor) regulates both the distal enhancer PBREM (phenobarbital-responsive enhancer module) and the proximal element OARE [OA (okadaic acid) response element] to synergistically up-regulate the endogenous CYP2B6 (where CYP is cytochrome P450) gene in HepG2 cells. Phenobarbital 88-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 275-279 16714371-1 2006 Cytochrome P450 (P450) enzymes metabolize the membrane lipid arachidonic acid to stable biologically active epoxides [eicosatrienoic acids (EETs)] and 20-hydroxyeicosatetraenoic acid (20-HETE). Arachidonic Acid 61-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 16714371-1 2006 Cytochrome P450 (P450) enzymes metabolize the membrane lipid arachidonic acid to stable biologically active epoxides [eicosatrienoic acids (EETs)] and 20-hydroxyeicosatetraenoic acid (20-HETE). Epoxy Compounds 108-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 16714371-1 2006 Cytochrome P450 (P450) enzymes metabolize the membrane lipid arachidonic acid to stable biologically active epoxides [eicosatrienoic acids (EETs)] and 20-hydroxyeicosatetraenoic acid (20-HETE). formic acid 118-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 16714371-1 2006 Cytochrome P450 (P450) enzymes metabolize the membrane lipid arachidonic acid to stable biologically active epoxides [eicosatrienoic acids (EETs)] and 20-hydroxyeicosatetraenoic acid (20-HETE). eets 140-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 16714371-1 2006 Cytochrome P450 (P450) enzymes metabolize the membrane lipid arachidonic acid to stable biologically active epoxides [eicosatrienoic acids (EETs)] and 20-hydroxyeicosatetraenoic acid (20-HETE). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 151-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 16714371-1 2006 Cytochrome P450 (P450) enzymes metabolize the membrane lipid arachidonic acid to stable biologically active epoxides [eicosatrienoic acids (EETs)] and 20-hydroxyeicosatetraenoic acid (20-HETE). 20-hydroxy-5,8,11,14-eicosatetraenoic acid 184-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 16714371-5 2006 Albendazole increased EETs by transcriptional induction of CYP1A5 and the others by combined induction of CYP1A5 and CYP2H, the avian orthologs of mammalian CYP1A2 and CYP2B, respectively. Albendazole 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 168-173 16581944-5 2006 Correlation analysis between the known P450 enzyme activities and the rate of the formation of endosulfan sulfate in the 14 human liver microsomes showed that alpha-endosulfan metabolism is significantly correlated with CYP2B6-mediated bupropion hydroxylation and CYP3A-mediated midazolam hydroxylation, and that beta-endosulfan metabolism is correlated with CYP3A activity. endosulfan sulfate 95-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-226 16769251-7 2006 The metabolism of many exogenous compounds including benzo(a)pyrene (BaP), pyrene, ethoxyresorufin, ethoxycoumarin and aniline is mediated by P450 enzymes in tissues of marine invertebrates. Benzo(a)pyrene 53-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 16769251-7 2006 The metabolism of many exogenous compounds including benzo(a)pyrene (BaP), pyrene, ethoxyresorufin, ethoxycoumarin and aniline is mediated by P450 enzymes in tissues of marine invertebrates. Benzo(a)pyrene 69-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 16769251-7 2006 The metabolism of many exogenous compounds including benzo(a)pyrene (BaP), pyrene, ethoxyresorufin, ethoxycoumarin and aniline is mediated by P450 enzymes in tissues of marine invertebrates. pyrene 61-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 16769251-7 2006 The metabolism of many exogenous compounds including benzo(a)pyrene (BaP), pyrene, ethoxyresorufin, ethoxycoumarin and aniline is mediated by P450 enzymes in tissues of marine invertebrates. ethoxyresorufin 83-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 16769251-7 2006 The metabolism of many exogenous compounds including benzo(a)pyrene (BaP), pyrene, ethoxyresorufin, ethoxycoumarin and aniline is mediated by P450 enzymes in tissues of marine invertebrates. 3-ethoxychromen-2-one 100-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 16769251-7 2006 The metabolism of many exogenous compounds including benzo(a)pyrene (BaP), pyrene, ethoxyresorufin, ethoxycoumarin and aniline is mediated by P450 enzymes in tissues of marine invertebrates. aniline 119-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 16581944-6 2006 The P450 isoform-selective inhibition study in human liver microsomes and the incubation study of cDNA-expressed enzymes also demonstrated that the stereoselective sulfonation of alpha-endosulfan is mediated by CYP2B6, CYP3A4, and CYP3A5, and that that of beta-endosulfan is transformed by CYP3A4 and CYP3A5. Endosulfan 256-271 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 211-217 16581944-8 2006 CYP2B6 enantioselectively metabolizes alpha-endosulfan, but not beta-endosulfan. Endosulfan 38-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 16543915-1 2006 Cytochrome P450 (CYP) enzyme 2B1 metabolizes the anticancer prodrug cyclophosphamide (CPA) to 4-hydroxy-CPA, which decomposes to the cytotoxic metabolites acrolein and phosphoramide mustard. Cyclophosphamide 68-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 16543915-1 2006 Cytochrome P450 (CYP) enzyme 2B1 metabolizes the anticancer prodrug cyclophosphamide (CPA) to 4-hydroxy-CPA, which decomposes to the cytotoxic metabolites acrolein and phosphoramide mustard. Cyclophosphamide 86-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 16543915-1 2006 Cytochrome P450 (CYP) enzyme 2B1 metabolizes the anticancer prodrug cyclophosphamide (CPA) to 4-hydroxy-CPA, which decomposes to the cytotoxic metabolites acrolein and phosphoramide mustard. 4-hydroxycyclophosphamide 94-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 16543915-1 2006 Cytochrome P450 (CYP) enzyme 2B1 metabolizes the anticancer prodrug cyclophosphamide (CPA) to 4-hydroxy-CPA, which decomposes to the cytotoxic metabolites acrolein and phosphoramide mustard. Acrolein 155-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 16543915-1 2006 Cytochrome P450 (CYP) enzyme 2B1 metabolizes the anticancer prodrug cyclophosphamide (CPA) to 4-hydroxy-CPA, which decomposes to the cytotoxic metabolites acrolein and phosphoramide mustard. phosphoramide 168-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 16867170-3 2006 The metabolism of pioglitazone by CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4 and CYP3A5 was investigated using human recombinant CYP isoforms. Pioglitazone 18-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 16581944-9 2006 These findings suggest that the CYP2B6 and CYP3A enzymes are major enzymes contributing to the stereoselective disposition of endosulfan. Endosulfan 126-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 16581944-5 2006 Correlation analysis between the known P450 enzyme activities and the rate of the formation of endosulfan sulfate in the 14 human liver microsomes showed that alpha-endosulfan metabolism is significantly correlated with CYP2B6-mediated bupropion hydroxylation and CYP3A-mediated midazolam hydroxylation, and that beta-endosulfan metabolism is correlated with CYP3A activity. Endosulfan 159-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-226 16581944-5 2006 Correlation analysis between the known P450 enzyme activities and the rate of the formation of endosulfan sulfate in the 14 human liver microsomes showed that alpha-endosulfan metabolism is significantly correlated with CYP2B6-mediated bupropion hydroxylation and CYP3A-mediated midazolam hydroxylation, and that beta-endosulfan metabolism is correlated with CYP3A activity. Bupropion 236-245 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-226 16581944-5 2006 Correlation analysis between the known P450 enzyme activities and the rate of the formation of endosulfan sulfate in the 14 human liver microsomes showed that alpha-endosulfan metabolism is significantly correlated with CYP2B6-mediated bupropion hydroxylation and CYP3A-mediated midazolam hydroxylation, and that beta-endosulfan metabolism is correlated with CYP3A activity. Midazolam 279-288 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-226 16581944-5 2006 Correlation analysis between the known P450 enzyme activities and the rate of the formation of endosulfan sulfate in the 14 human liver microsomes showed that alpha-endosulfan metabolism is significantly correlated with CYP2B6-mediated bupropion hydroxylation and CYP3A-mediated midazolam hydroxylation, and that beta-endosulfan metabolism is correlated with CYP3A activity. Endosulfan 313-328 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-226 16581944-6 2006 The P450 isoform-selective inhibition study in human liver microsomes and the incubation study of cDNA-expressed enzymes also demonstrated that the stereoselective sulfonation of alpha-endosulfan is mediated by CYP2B6, CYP3A4, and CYP3A5, and that that of beta-endosulfan is transformed by CYP3A4 and CYP3A5. Endosulfan 179-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 211-217 16639745-0 2006 Multiple molecular dynamics simulations of human p450 monooxygenase CYP2C9: the molecular basis of substrate binding and regioselectivity toward warfarin. Warfarin 145-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 16608219-0 2006 Phytoremediation of the herbicides atrazine and metolachlor by transgenic rice plants expressing human CYP1A1, CYP2B6, and CYP2C19. Atrazine 35-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 16855167-0 2006 DHEA metabolism in arthritis: a role for the p450 enzyme Cyp7b at the immune-endocrine crossroad. Dehydroepiandrosterone 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-49 16855167-3 2006 The described immuno-stimulating effects of DHEA may be due to the conversion of DHEA into 7alpha-hydroxy-DHEA (7alpha-OH-DHEA) by the activity of the p450 enzyme, Cyp7b. Dehydroepiandrosterone 44-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-155 16855167-3 2006 The described immuno-stimulating effects of DHEA may be due to the conversion of DHEA into 7alpha-hydroxy-DHEA (7alpha-OH-DHEA) by the activity of the p450 enzyme, Cyp7b. Dehydroepiandrosterone 81-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-155 16855167-3 2006 The described immuno-stimulating effects of DHEA may be due to the conversion of DHEA into 7alpha-hydroxy-DHEA (7alpha-OH-DHEA) by the activity of the p450 enzyme, Cyp7b. 7-hydroxydehydroepiandrosterone 91-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-155 16855167-3 2006 The described immuno-stimulating effects of DHEA may be due to the conversion of DHEA into 7alpha-hydroxy-DHEA (7alpha-OH-DHEA) by the activity of the p450 enzyme, Cyp7b. 7alpha-Hydroxydehydroepiandrosterone 112-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-155 16855167-16 2006 The role of the p450 enzyme Cyp7b in DHEA metabolism and its relevance in the arthritic process will be discussed. Dehydroepiandrosterone 37-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 16545970-8 2006 The enzymatic decomposition of V-PYRRO/NO was also catalyzed, albeit at lower rates, by CYP2A6 and CYP2B6. v-pyrro 31-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 16607661-4 2006 The significant enhancement of the scattering intensity of QDs observed upon conjugation with the P450(BSbeta) due to the refractive-index increment and the systematic variation in zeta potential resulting from charge neutralization of the anionic QDs by the cationic histidine-tagged P450(BSbeta) have been used for stoichiometry determination. Histidine 268-277 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-110 16608219-0 2006 Phytoremediation of the herbicides atrazine and metolachlor by transgenic rice plants expressing human CYP1A1, CYP2B6, and CYP2C19. metolachlor 48-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 16608165-12 2006 Our present findings indicate that the oxidase activity of P450 (i.e., formation of ROS) represents a mechanism of deboronation. Reactive Oxygen Species 84-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-63 16854777-0 2006 Stereoselectivity in metabolism of ifosfamide by CYP3A4 and CYP2B6. Ifosfamide 35-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 16415119-7 2006 A monoclonal antibody to CYP2B6 and the CYP3A inhibitor ketoconazole substantially inhibited R-138727 formation, whereas inhibitors of CYP2C9 (sulfaphenazole) and CYP2C19 (omeprazole) did not. Sulfaphenazole 143-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 16608165-0 2006 Oxidative deboronation of the peptide boronic acid proteasome inhibitor bortezomib: contributions from reactive oxygen species in this novel cytochrome P450 reaction. Bortezomib 72-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-156 16608165-0 2006 Oxidative deboronation of the peptide boronic acid proteasome inhibitor bortezomib: contributions from reactive oxygen species in this novel cytochrome P450 reaction. Reactive Oxygen Species 103-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-156 16608165-6 2006 Use of 18O-labeled oxygen under controlled atmospheres confirmed an oxidative mechanism in the P450-mediated deboronation of 1, as 18O was found incorporated in both M1 and M2. 18o 7-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 16608165-6 2006 Use of 18O-labeled oxygen under controlled atmospheres confirmed an oxidative mechanism in the P450-mediated deboronation of 1, as 18O was found incorporated in both M1 and M2. Oxygen 19-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 16608165-6 2006 Use of 18O-labeled oxygen under controlled atmospheres confirmed an oxidative mechanism in the P450-mediated deboronation of 1, as 18O was found incorporated in both M1 and M2. 18o 131-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 16608165-7 2006 Chemically generated reactive oxygen species (ROS), such as those generated as byproducts during P450 catalysis, were also found to deboronate 1 resulting in the formation of M1 and M2. Reactive Oxygen Species 21-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-101 16608165-7 2006 Chemically generated reactive oxygen species (ROS), such as those generated as byproducts during P450 catalysis, were also found to deboronate 1 resulting in the formation of M1 and M2. Reactive Oxygen Species 46-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-101 16608165-8 2006 Known to undergo efficient redox cycling, P450 2E1 was found to catalyze the deboronation of 1 predominantly to the carbinolamide metabolites M1 and M2, as well as to a pair of peroxycarbinolamides, 2 and 3. carbinolamide 116-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 16608165-8 2006 Known to undergo efficient redox cycling, P450 2E1 was found to catalyze the deboronation of 1 predominantly to the carbinolamide metabolites M1 and M2, as well as to a pair of peroxycarbinolamides, 2 and 3. peroxycarbinolamides 177-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 16608165-9 2006 The presence of superoxide dismutase (SOD) and catalase prevented the deboronation of 1, thus, supporting the involvement of ROS in the P450 2E1-catalyzed deboronation reaction. Reactive Oxygen Species 125-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 16415119-7 2006 A monoclonal antibody to CYP2B6 and the CYP3A inhibitor ketoconazole substantially inhibited R-138727 formation, whereas inhibitors of CYP2C9 (sulfaphenazole) and CYP2C19 (omeprazole) did not. R-138727 93-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 16415119-7 2006 A monoclonal antibody to CYP2B6 and the CYP3A inhibitor ketoconazole substantially inhibited R-138727 formation, whereas inhibitors of CYP2C9 (sulfaphenazole) and CYP2C19 (omeprazole) did not. Omeprazole 172-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 16415125-6 2006 Using either cholate method, more than 85% of the P450 was physically incorporated into the phospholipid vesicles, whereas less than 40% of the P450 was physically incorporated into the phospholipid vesicles using the SRS. Phospholipids 92-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 16415125-11 2006 Both cholate methods resulted in the loss of a proportion of spectrally detectable carbon monoxyferrous P450, resulting from incubation of the proteins with detergent. Cholates 5-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-108 16510191-0 2006 The status of high-valent metal oxo complexes in the P450 cytochromes. metal oxo 26-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 16506047-1 2006 OBJECTIVE: Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of artemisinin drugs, a novel series of antimalarials. artemisinin 73-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-30 16506047-1 2006 OBJECTIVE: Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of artemisinin drugs, a novel series of antimalarials. artemisinin 73-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 16510191-1 2006 The oxidative prowess of the P450 cytochromes in physiological reactions is attributed to the production of a high-valent iron-oxo complex, or Compound I intermediate, in the reaction cycle. iron-oxo 122-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 16510191-5 2006 Structural and spectroscopic studies on manganese reconstituted P450, and its corresponding oxo-complex, are presented. Manganese 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 16550168-6 2006 This includes also some anticancer drugs like tamoxifen and cyclophosphamide metabolized by CYP2D6, CYP2C19 and CYP2B6. Tamoxifen 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-118 16550168-6 2006 This includes also some anticancer drugs like tamoxifen and cyclophosphamide metabolized by CYP2D6, CYP2C19 and CYP2B6. Cyclophosphamide 60-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-118 21638641-1 2006 Cytochrome P450 (P450) enzymes include a family of related enzymes that are involved in metabolism of vitamins, steroids, fatty acids, and other chemicals. Fatty Acids 122-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 21638641-1 2006 Cytochrome P450 (P450) enzymes include a family of related enzymes that are involved in metabolism of vitamins, steroids, fatty acids, and other chemicals. Steroids 112-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 16373351-0 2006 Structure of microsomal cytochrome P450 2B4 complexed with the antifungal drug bifonazole: insight into P450 conformational plasticity and membrane interaction. bifonazole 79-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 16373351-0 2006 Structure of microsomal cytochrome P450 2B4 complexed with the antifungal drug bifonazole: insight into P450 conformational plasticity and membrane interaction. bifonazole 79-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-108 16484569-0 2006 In vitro interactions of water-soluble garlic components with human cytochromes p450. Water 25-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 16309716-0 2006 CYP2B6 is expressed in African Green monkey brain and is induced by chronic nicotine treatment. Nicotine 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 16309716-1 2006 CYP2B6 is a drug-metabolizing enzyme expressed in human tissues that can activate bupropion (a smoking cessation drug) and tobacco smoke nitrosamines and can inactivate drugs such as nicotine. Bupropion 82-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 16309716-1 2006 CYP2B6 is a drug-metabolizing enzyme expressed in human tissues that can activate bupropion (a smoking cessation drug) and tobacco smoke nitrosamines and can inactivate drugs such as nicotine. Nitrosamines 137-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 16309716-1 2006 CYP2B6 is a drug-metabolizing enzyme expressed in human tissues that can activate bupropion (a smoking cessation drug) and tobacco smoke nitrosamines and can inactivate drugs such as nicotine. Nicotine 183-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 16309716-3 2006 We investigated the basal expression and the effect of chronic nicotine treatment on CYP2B6 protein in African Green monkey (Cercopithecus aethiops) brain. Nicotine 63-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 16309716-7 2006 Chronic nicotine treatment induced CYP2B6 expression in specific cells such as astrocytes and neurons in the frontal cortex, caudate, thalamus and hippocampus. Nicotine 8-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 16309716-10 2006 In conclusion, CYP2B6 protein is expressed in specific cells in monkey brain and is induced by chronic nicotine treatment which may impact central metabolism of CYP2B6 substrates such as bupropion and nicotine. Nicotine 103-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 16309716-10 2006 In conclusion, CYP2B6 protein is expressed in specific cells in monkey brain and is induced by chronic nicotine treatment which may impact central metabolism of CYP2B6 substrates such as bupropion and nicotine. Nicotine 103-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 16309716-10 2006 In conclusion, CYP2B6 protein is expressed in specific cells in monkey brain and is induced by chronic nicotine treatment which may impact central metabolism of CYP2B6 substrates such as bupropion and nicotine. Bupropion 187-196 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 16309716-10 2006 In conclusion, CYP2B6 protein is expressed in specific cells in monkey brain and is induced by chronic nicotine treatment which may impact central metabolism of CYP2B6 substrates such as bupropion and nicotine. Bupropion 187-196 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 16309716-10 2006 In conclusion, CYP2B6 protein is expressed in specific cells in monkey brain and is induced by chronic nicotine treatment which may impact central metabolism of CYP2B6 substrates such as bupropion and nicotine. Nicotine 201-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 16546968-1 2006 Gene therapy using the prodrug-activating enzyme P450 2B6 has shown substantial promise in preclinical and initial clinical studies with the P450 prodrugs cyclophosphamide and ifosfamide. Cyclophosphamide 155-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 16546968-1 2006 Gene therapy using the prodrug-activating enzyme P450 2B6 has shown substantial promise in preclinical and initial clinical studies with the P450 prodrugs cyclophosphamide and ifosfamide. Cyclophosphamide 155-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 16546968-1 2006 Gene therapy using the prodrug-activating enzyme P450 2B6 has shown substantial promise in preclinical and initial clinical studies with the P450 prodrugs cyclophosphamide and ifosfamide. Ifosfamide 176-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 16546968-1 2006 Gene therapy using the prodrug-activating enzyme P450 2B6 has shown substantial promise in preclinical and initial clinical studies with the P450 prodrugs cyclophosphamide and ifosfamide. Ifosfamide 176-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 16546968-2 2006 We sought to optimize this therapy using the canine P450 enzyme 2B11, which activates cyclophosphamide and ifosfamide with Km of 80 to 160 micromol/L, approximately 10- to 20-fold lower than the Km of P450 2B6. Cyclophosphamide 86-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 16546968-2 2006 We sought to optimize this therapy using the canine P450 enzyme 2B11, which activates cyclophosphamide and ifosfamide with Km of 80 to 160 micromol/L, approximately 10- to 20-fold lower than the Km of P450 2B6. Ifosfamide 107-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 16546968-2 2006 We sought to optimize this therapy using the canine P450 enzyme 2B11, which activates cyclophosphamide and ifosfamide with Km of 80 to 160 micromol/L, approximately 10- to 20-fold lower than the Km of P450 2B6. Ifosfamide 107-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 201-205 16546968-3 2006 Retrovirus encoding a P450 2B11-internal ribosome entry signal-P450 reductase expression cassette induced marked cyclophosphamide and ifosfamide cytotoxicity toward 9L gliosarcoma cells and exhibited an impressive bystander killing effect at micromolar prodrug concentrations, where P450 2B6 displayed low activity. Cyclophosphamide 113-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 16546968-3 2006 Retrovirus encoding a P450 2B11-internal ribosome entry signal-P450 reductase expression cassette induced marked cyclophosphamide and ifosfamide cytotoxicity toward 9L gliosarcoma cells and exhibited an impressive bystander killing effect at micromolar prodrug concentrations, where P450 2B6 displayed low activity. Cyclophosphamide 113-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 16546968-3 2006 Retrovirus encoding a P450 2B11-internal ribosome entry signal-P450 reductase expression cassette induced marked cyclophosphamide and ifosfamide cytotoxicity toward 9L gliosarcoma cells and exhibited an impressive bystander killing effect at micromolar prodrug concentrations, where P450 2B6 displayed low activity. Cyclophosphamide 113-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 16546968-3 2006 Retrovirus encoding a P450 2B11-internal ribosome entry signal-P450 reductase expression cassette induced marked cyclophosphamide and ifosfamide cytotoxicity toward 9L gliosarcoma cells and exhibited an impressive bystander killing effect at micromolar prodrug concentrations, where P450 2B6 displayed low activity. Ifosfamide 134-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 16546968-3 2006 Retrovirus encoding a P450 2B11-internal ribosome entry signal-P450 reductase expression cassette induced marked cyclophosphamide and ifosfamide cytotoxicity toward 9L gliosarcoma cells and exhibited an impressive bystander killing effect at micromolar prodrug concentrations, where P450 2B6 displayed low activity. Ifosfamide 134-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 16546968-3 2006 Retrovirus encoding a P450 2B11-internal ribosome entry signal-P450 reductase expression cassette induced marked cyclophosphamide and ifosfamide cytotoxicity toward 9L gliosarcoma cells and exhibited an impressive bystander killing effect at micromolar prodrug concentrations, where P450 2B6 displayed low activity. Ifosfamide 134-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 16546968-4 2006 Adeno-2B11, a replication-defective, E1/E3 region-deleted adenovirus engineered to coexpress P450 2B11 and P450 reductase, dramatically increased tumor cell-catalyzed cyclophosphamide 4-hydroxylation and cytotoxicity compared with Adeno-2B6 and effected strong bystander killing at low (20 micromol/L) cyclophosphamide concentrations. Cyclophosphamide 167-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 16546968-4 2006 Adeno-2B11, a replication-defective, E1/E3 region-deleted adenovirus engineered to coexpress P450 2B11 and P450 reductase, dramatically increased tumor cell-catalyzed cyclophosphamide 4-hydroxylation and cytotoxicity compared with Adeno-2B6 and effected strong bystander killing at low (20 micromol/L) cyclophosphamide concentrations. Cyclophosphamide 167-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-111 16546968-4 2006 Adeno-2B11, a replication-defective, E1/E3 region-deleted adenovirus engineered to coexpress P450 2B11 and P450 reductase, dramatically increased tumor cell-catalyzed cyclophosphamide 4-hydroxylation and cytotoxicity compared with Adeno-2B6 and effected strong bystander killing at low (20 micromol/L) cyclophosphamide concentrations. Cyclophosphamide 302-318 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 16546968-9 2006 Thus, P450 gene-directed enzyme prodrug therapy can be greatly improved by using the low Km P450 enzyme 2B11, which catalyzes intratumoral activation of cyclophosphamide and ifosfamide at pharmacologically relevant drug concentrations. Cyclophosphamide 153-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-10 16546968-9 2006 Thus, P450 gene-directed enzyme prodrug therapy can be greatly improved by using the low Km P450 enzyme 2B11, which catalyzes intratumoral activation of cyclophosphamide and ifosfamide at pharmacologically relevant drug concentrations. Cyclophosphamide 153-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 16546968-9 2006 Thus, P450 gene-directed enzyme prodrug therapy can be greatly improved by using the low Km P450 enzyme 2B11, which catalyzes intratumoral activation of cyclophosphamide and ifosfamide at pharmacologically relevant drug concentrations. Ifosfamide 174-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-10 16546968-9 2006 Thus, P450 gene-directed enzyme prodrug therapy can be greatly improved by using the low Km P450 enzyme 2B11, which catalyzes intratumoral activation of cyclophosphamide and ifosfamide at pharmacologically relevant drug concentrations. Ifosfamide 174-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 16309716-10 2006 In conclusion, CYP2B6 protein is expressed in specific cells in monkey brain and is induced by chronic nicotine treatment which may impact central metabolism of CYP2B6 substrates such as bupropion and nicotine. Nicotine 201-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 16293390-0 2006 Activation of oxazaphosphorines by cytochrome P450: application to gene-directed enzyme prodrug therapy for cancer. oxazaphosphorines 14-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-50 16495778-1 2006 The non-nucleoside reverse transcriptase inhibitor efavirenz is mainly metabolised by the polymorphic cytochrome P450 enzyme CYP2B6. efavirenz 51-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 16495778-2 2006 Genomic DNA from four subjects in a group of 51 patients being treated with efavirenz and having surprisingly high plasma concentrations were screened by direct sequencing for mutations in the CYP2B6 gene. efavirenz 76-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-199 16495778-9 2006 The steady-state level of efavirenz was significantly higher in the five carriers of CYP2B6*16, being of African origin, compared to the other patients. efavirenz 26-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 16495778-10 2006 Higher efavirenz concentrations were also seen in carriers of 516G>T (CYP2B6*6 and CYP2B6*9). efavirenz 7-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 16495778-10 2006 Higher efavirenz concentrations were also seen in carriers of 516G>T (CYP2B6*6 and CYP2B6*9). efavirenz 7-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 16495778-11 2006 In conclusion, a novel CYP2B6*16 allele causing less expression of the corresponding enzyme was identified and found to influence the metabolism of efavirenz in vivo, a finding that is of potential impact for anti-HIV therapy in black populations. efavirenz 148-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 16498227-4 2006 We highlight the data linking such low-level Cd intake with tubular injury, altered abundance of cytochromes P450 (CYPs) in the kidney and an expression of a hypertensive phenotype. Cadmium 45-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 16293390-1 2006 Cancer chemotherapeutic prodrugs, such as the oxazaphosphorines cyclophosphamide and ifosfamide, are metabolized by liver cytochrome P450 enzymes to yield therapeutically active, cytotoxic metabolites. oxazaphosphorines 46-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 16293390-1 2006 Cancer chemotherapeutic prodrugs, such as the oxazaphosphorines cyclophosphamide and ifosfamide, are metabolized by liver cytochrome P450 enzymes to yield therapeutically active, cytotoxic metabolites. Cyclophosphamide 64-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 16293390-1 2006 Cancer chemotherapeutic prodrugs, such as the oxazaphosphorines cyclophosphamide and ifosfamide, are metabolized by liver cytochrome P450 enzymes to yield therapeutically active, cytotoxic metabolites. Ifosfamide 85-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 16293390-3 2006 This problem can, in part, be circumvented by implementation of cytochrome P450 gene-directed enzyme prodrug therapy (P450 GDEPT), a prodrug activation strategy for cancer treatment that augments tumor cell exposure to cytotoxic drug metabolites generated locally by a prodrug-activating cytochrome P450 enzyme. gdept 123-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 16293390-3 2006 This problem can, in part, be circumvented by implementation of cytochrome P450 gene-directed enzyme prodrug therapy (P450 GDEPT), a prodrug activation strategy for cancer treatment that augments tumor cell exposure to cytotoxic drug metabolites generated locally by a prodrug-activating cytochrome P450 enzyme. gdept 123-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 16293390-3 2006 This problem can, in part, be circumvented by implementation of cytochrome P450 gene-directed enzyme prodrug therapy (P450 GDEPT), a prodrug activation strategy for cancer treatment that augments tumor cell exposure to cytotoxic drug metabolites generated locally by a prodrug-activating cytochrome P450 enzyme. gdept 123-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 16272404-2 2006 The purpose of this in vitro study was to identify the phase I metabolites, potential species differences in metabolism, and the cytochromes P450 (P450s) involved in the phase I metabolism of S4 using 14C-S4, recombinant P450s, and other liver enzyme preparations from human, rat, and dog. Carbon-14 201-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 16460014-3 2006 Here, we use UV/vis and heteronuclear nuclear magnetic resonance (NMR) spectroscopic techniques to study the mechanism and thermodynamics of the binding of two 9-aminophenanthrene (9-AP) and testosterone (TST) molecules to the erythromycin-metabolizing bacterial P450(eryF). 9-aminophenanthrene 160-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 263-273 16460014-3 2006 Here, we use UV/vis and heteronuclear nuclear magnetic resonance (NMR) spectroscopic techniques to study the mechanism and thermodynamics of the binding of two 9-aminophenanthrene (9-AP) and testosterone (TST) molecules to the erythromycin-metabolizing bacterial P450(eryF). 9-aminophenanthrene 181-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 263-273 16460014-3 2006 Here, we use UV/vis and heteronuclear nuclear magnetic resonance (NMR) spectroscopic techniques to study the mechanism and thermodynamics of the binding of two 9-aminophenanthrene (9-AP) and testosterone (TST) molecules to the erythromycin-metabolizing bacterial P450(eryF). Testosterone 191-203 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 263-273 16389533-3 2006 METHODS: Rabeprazole-thioether was incubated with human liver microsomes and several recombinant cytochrome P450 (CYP) enzymes (CYPs 1A2, 2A6, 2B6, 2C9, 2C19, 2D6, 2E1, and 3A4). rabeprazole-thioether 9-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 16389533-7 2006 The intrinsic clearance (V(max ) /K(m )) for the oxidation by CYP3A4 of (R)-rabeprazole was 3.5-fold higher than that for the (S)-enantiomer (81 nl/min/pmol of P450 vs. 23 nl/min/pmol of P450). dexrabeprazole 72-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 160-164 16389533-7 2006 The intrinsic clearance (V(max ) /K(m )) for the oxidation by CYP3A4 of (R)-rabeprazole was 3.5-fold higher than that for the (S)-enantiomer (81 nl/min/pmol of P450 vs. 23 nl/min/pmol of P450). dexrabeprazole 72-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-191 16389533-9 2006 The mean K(m ) and V(max ) values of desmethylrabeprazole-thioether formation for CYP2C19 were 5.1 microM and 600 pmol/min/nmol of P450, respectively, whereas those for CYP2D6 were 15.1 microM and 736 pmol/min/nmol of P450, respectively. desmethylrabeprazole-thioether 37-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-135 16389533-9 2006 The mean K(m ) and V(max ) values of desmethylrabeprazole-thioether formation for CYP2C19 were 5.1 microM and 600 pmol/min/nmol of P450, respectively, whereas those for CYP2D6 were 15.1 microM and 736 pmol/min/nmol of P450, respectively. desmethylrabeprazole-thioether 37-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 218-222 16443151-5 2006 However, most of the biological effects of arachidonic acid are attributable to its metabolism by mainly three different groups of enzymes: cytochromes P450, cyclooxygenases, and lipoxygenases. Arachidonic Acid 43-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-192 16460014-4 2006 UV/vis absorbance spectra of P450(eryF) demonstrated that binding occurs with apparent negative homotropic cooperativity for TST and positive homotropic cooperativity for 9-AP with Hill-equation-derived dissociation constants of K(S) = 4 and 200 microM, respectively. 9-aminophenanthrene 171-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-39 16460014-5 2006 The broadening and shifting observed in the 2D-{1H,15N}-HSQC-monitored titrations of 15N-Phe-labeled P450(eryF) with 9-AP and TST indicated binding on intermediate and fast chemical exchange time scales, respectively, which was consistent with the Hill-equation-derived K(S) values for these two ligands. Hydrogen 48-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-111 16460014-5 2006 The broadening and shifting observed in the 2D-{1H,15N}-HSQC-monitored titrations of 15N-Phe-labeled P450(eryF) with 9-AP and TST indicated binding on intermediate and fast chemical exchange time scales, respectively, which was consistent with the Hill-equation-derived K(S) values for these two ligands. 15n 51-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-111 16460014-5 2006 The broadening and shifting observed in the 2D-{1H,15N}-HSQC-monitored titrations of 15N-Phe-labeled P450(eryF) with 9-AP and TST indicated binding on intermediate and fast chemical exchange time scales, respectively, which was consistent with the Hill-equation-derived K(S) values for these two ligands. 15n 85-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-111 16460014-5 2006 The broadening and shifting observed in the 2D-{1H,15N}-HSQC-monitored titrations of 15N-Phe-labeled P450(eryF) with 9-AP and TST indicated binding on intermediate and fast chemical exchange time scales, respectively, which was consistent with the Hill-equation-derived K(S) values for these two ligands. Phenylalanine 89-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-111 16392089-11 2006 CONCLUSIONS: The CYP2B6 position 516 TT genotype or a prolonged measured elimination half-life may predict increased risk of developing drug resistance among patients who discontinue efavirenz-containing regimens. efavirenz 183-192 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 16272404-2 2006 The purpose of this in vitro study was to identify the phase I metabolites, potential species differences in metabolism, and the cytochromes P450 (P450s) involved in the phase I metabolism of S4 using 14C-S4, recombinant P450s, and other liver enzyme preparations from human, rat, and dog. Carbon-14 201-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-152 16360114-5 2006 The purified P450 was in the low-spin iron state, and the spin equilibrium was not perturbed by any of the potential substrates vitamin D(3), 1alpha- or 25-hydroxy vitamin D(3), or cholesterol. Iron 38-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 16214851-2 2006 Various bioactivating enzymes, such as cytochromes P450 and myeloperoxidase, have been shown to be capable of catalyzing the N-oxidation of these compounds. Nitrogen 125-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-75 16413243-2 2006 METHODS: Candidate pharmacogenetic modulators were polymorphisms reported to be of functional consequence and therefore potentially important for metabolism, distribution, or pharmacodynamic action of levomethadone, consisting of genes coding for cytochrome P450 (CYP) 2B6 and 3A, as well as 1A2, 2C8, 2C9, 2C19, and 2D6, for P-glycoprotein (ABCB1), and for mu-opioid receptors (OPRM1). Levomethadone 201-214 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 247-279 16243959-0 2006 In vitro metabolism of naphthalene by human liver microsomal cytochrome P450 enzymes. naphthalene 23-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-76 16243959-6 2006 P450 isoform screening of naphthalene metabolism identified CYP1A2 as the most efficient isoform for producing dihydrodiol and 1-naphthol, and CYP3A4 as the most effective for 2-naphthol production. trans-1,2-dihydro-1,2-naphthalenediol 111-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 16243959-6 2006 P450 isoform screening of naphthalene metabolism identified CYP1A2 as the most efficient isoform for producing dihydrodiol and 1-naphthol, and CYP3A4 as the most effective for 2-naphthol production. 1-naphthol 127-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 16243959-6 2006 P450 isoform screening of naphthalene metabolism identified CYP1A2 as the most efficient isoform for producing dihydrodiol and 1-naphthol, and CYP3A4 as the most effective for 2-naphthol production. 2-naphthol 176-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 16243959-3 2006 The current studies were performed to characterize naphthalene metabolism by human cytochromes P450. naphthalene 51-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 16243959-9 2006 A series of human p450 isoforms was used to further explore the metabolism of dihydrodiol and 1-naphthol. trans-1,2-dihydro-1,2-naphthalenediol 78-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-22 16243959-6 2006 P450 isoform screening of naphthalene metabolism identified CYP1A2 as the most efficient isoform for producing dihydrodiol and 1-naphthol, and CYP3A4 as the most effective for 2-naphthol production. naphthalene 26-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 16243959-9 2006 A series of human p450 isoforms was used to further explore the metabolism of dihydrodiol and 1-naphthol. 1-naphthol 94-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-22 16243959-11 2006 Dihydrodiol was metabolized by P450 isoforms to three minor unidentified metabolites with CYP3A4 and CYP2A6 having the greatest activity toward this substrate. trans-1,2-dihydro-1,2-naphthalenediol 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 16719384-1 2006 Testosterone and other steroid hormones have been studied as prototypic examples of endogenous substrates for hepatic cytochrome P450 (P450) enzymes. Testosterone 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-140 16243959-12 2006 The metabolism of dihydrodiol by P450 isoforms was lower than that of 1-naphthol. trans-1,2-dihydro-1,2-naphthalenediol 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 16243959-12 2006 The metabolism of dihydrodiol by P450 isoforms was lower than that of 1-naphthol. 1-naphthol 70-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 16243959-13 2006 These studies identify primary and secondary metabolites of naphthalene produced by pHLMs and P450 isoforms. naphthalene 60-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-98 16183265-3 2006 The pharmacokinetics of CPA and its active metabolite were related to the genotype of CYP2B6, CYP2C9 and CYP2C19. Cyclophosphamide 24-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 16183265-13 2006 In the population analysis, the CYP2B6 G516T variant allele contribution to CPA clearance was about twice as the contribution from the wild type gene while the genotype of CYP2C9 and CYP2C19 did not influence clearance. Cyclophosphamide 76-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 16183265-15 2006 CONCLUSION: This study demonstrates for the first time that the presence of the CYP2B6 G516T mutation increases the rate of 4-OH-CPA formation in patients with hematological malignancies. 4-oh-cpa 124-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 16484082-0 2006 Biotransformation of metamitron by human p450 expressed in transgenic tobacco cell cultures. metamitron 21-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 16484082-1 2006 In the present investigation, the oxidative metabolism of 14C-labeled metamitron was examined in plant cell cultures of tobacco overexpressing human P450 enzymes CYP1A1 or CYP1A2; special interest was in the aromatic hydroxylation of the herbicide. Carbon-14 58-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-153 16484082-1 2006 In the present investigation, the oxidative metabolism of 14C-labeled metamitron was examined in plant cell cultures of tobacco overexpressing human P450 enzymes CYP1A1 or CYP1A2; special interest was in the aromatic hydroxylation of the herbicide. metamitron 70-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-153 16484082-6 2006 In a large-scale study (up to 400 microg per assay) with the transgenic culture expressing CYP1A2, the high efficiency of this P450 system toward metamitron was demonstrated: turnover of the xenobiotic was almost complete with 400 microg. metamitron 146-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 27699667-1 2006 Testosterone and other steroid hormones have been studied as prototypic examples of endogenous substrates for hepatic cytochrome P450 (P450) enzymes. Testosterone 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-140 27699667-1 2006 Testosterone and other steroid hormones have been studied as prototypic examples of endogenous substrates for hepatic cytochrome P450 (P450) enzymes. Steroids 23-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-140 27699679-1 2006 Cytochrome P450 (P450) is a superfamily of individual monooxygenase enzymes that metabolize structurally diverse xenochemicals, including many clinically useful drugs and foreign chemicals widespread in the environment. xenochemicals 113-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 16719375-1 2006 Cytochrome P450 (P450) is a superfamily of individual monooxygenase enzymes that metabolize structurally diverse xenochemicals, including many clinically useful drugs and foreign chemicals widespread in the environment. xenochemicals 113-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 16719376-1 2006 Cytochrome P450 (P450) enzymes belonging to the CYP1 family are highly inducible by polycyclic aromatic hydrocarbons and other environmental chemicals and play a major role in the metabolism of many foreign chemicals and endogenous substances. Polycyclic Aromatic Hydrocarbons 84-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 16719378-0 2006 Determination of CYP2B6 component of 7-ethoxy-4-trifluoromethylcoumarin O-deethylation activity in human liver microsomes. 7-ethoxy-4-trifluoromethylcoumarin 37-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 16719378-1 2006 The cytochrome P450 enzyme CYP2B6 plays an important role in the metabolism of structurally diverse drugs, including the anticancer drug cyclophosphamide, and may be an important determinant of clinical responses to these agents. Cyclophosphamide 137-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 16719378-2 2006 A spectrofluorometric method is described for the determination of CYP2B6-catalyzed 7-ethoxy-4-trifluoromethylcoumarin O-deethylation activity in human liver microsomes. 7-ethoxy-4-trifluoromethylcoumarin 84-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 16719378-3 2006 The specificity of this method for CYP2B6 is increased by the use of inhibitory antibodies to CYP1A2, CYP2C, and CYP2E1, which block the contributions of these higher-K(m) enzymes to human liver microsomal metabolism of 7-ethoxy-4-trifluoromethylcoumarin. 7-ethoxy-4-trifluoromethylcoumarin 220-254 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 16719384-1 2006 Testosterone and other steroid hormones have been studied as prototypic examples of endogenous substrates for hepatic cytochrome P450 (P450) enzymes. Steroids 23-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-140 16125881-4 2005 The subjects" genetic polymorphisms in the genes that encode the styrene-metabolizing enzymes CYP2E1, CYP2B6, EPHX1, GSTM1, GSTT1 and GSTP1 were determined. Styrene 65-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 16338275-1 2005 BACKGROUND AND OBJECTIVE: Recent in vitro studies have suggested an important role of cytochrome P450 (CYP) 2B6 and CYP2C19 in methadone metabolism. Methadone 127-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-111 16246642-3 2005 Bupropion is used as a probe for the activity of the CYP2B6 isoenzyme of the P450 family of enzymes in man. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 16338275-2 2005 We aimed to determine the influence of CYP2B6, CYP2C9, and CYP2C19 genetic polymorphism on methadone pharmacokinetics and on the response to treatment. Methadone 91-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 16338275-6 2005 RESULTS: CYP2B6 genotype influences (S)-methadone and, to a lesser extent, (R)-methadone plasma levels, with the median trough (S)-methadone plasma levels being 105, 122, and 209 ng . Methadone 36-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 16338275-6 2005 RESULTS: CYP2B6 genotype influences (S)-methadone and, to a lesser extent, (R)-methadone plasma levels, with the median trough (S)-methadone plasma levels being 105, 122, and 209 ng . Methadone 75-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 16338275-6 2005 RESULTS: CYP2B6 genotype influences (S)-methadone and, to a lesser extent, (R)-methadone plasma levels, with the median trough (S)-methadone plasma levels being 105, 122, and 209 ng . Methadone 40-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 16338275-13 2005 CONCLUSION: Although CYP2B6 influences (S)-methadone plasma levels, given that only (R)-methadone contributes to the opioid effect of this drug, a major influence of CYP2B6 genotype on response to treatment is unlikely and has not been shown in this study. Methadone 39-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 16135656-11 2005 These results as well as the inhibition analyses suggested that CYP2A6 and CYP2B6 would significantly contribute to the nicotine N-demethylation at low and high substrate concentrations, respectively. Nicotine 120-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 16135659-7 2005 All 23 single assays were validated by assessing the effects (induction or repression) of known inducers (ethanol, 3-methylcholanthrene, rifampicin, dexamethasone, phenobarbital) on P450 expression in human primary hepatocytes. Ethanol 106-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-186 16135656-0 2005 CYP2A6 AND CYP2B6 are involved in nornicotine formation from nicotine in humans: interindividual differences in these contributions. nornicotine 34-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 16135656-0 2005 CYP2A6 AND CYP2B6 are involved in nornicotine formation from nicotine in humans: interindividual differences in these contributions. Nicotine 37-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 16135659-7 2005 All 23 single assays were validated by assessing the effects (induction or repression) of known inducers (ethanol, 3-methylcholanthrene, rifampicin, dexamethasone, phenobarbital) on P450 expression in human primary hepatocytes. Methylcholanthrene 115-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-186 16135656-10 2005 The nicotine N-demethylase activity at 100 microM nicotine was significantly correlated with the CYP2B6 contents (r = 0.677, p < 0.05) and S-mephenytoin N-demethylase activities (r = 0.740, p < 0.005). Nicotine 4-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 16135659-7 2005 All 23 single assays were validated by assessing the effects (induction or repression) of known inducers (ethanol, 3-methylcholanthrene, rifampicin, dexamethasone, phenobarbital) on P450 expression in human primary hepatocytes. Rifampin 137-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-186 16135659-7 2005 All 23 single assays were validated by assessing the effects (induction or repression) of known inducers (ethanol, 3-methylcholanthrene, rifampicin, dexamethasone, phenobarbital) on P450 expression in human primary hepatocytes. Dexamethasone 149-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-186 16135659-7 2005 All 23 single assays were validated by assessing the effects (induction or repression) of known inducers (ethanol, 3-methylcholanthrene, rifampicin, dexamethasone, phenobarbital) on P450 expression in human primary hepatocytes. Phenobarbital 164-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 182-186 16415532-0 2005 Localization of rat cytochrome P450 in various tissues and comparison of arachidonic acid metabolism by rat P450 with that by human P450 orthologs. Arachidonic Acid 73-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 16379671-6 2005 Delta(3)-carene-10-ol was formed by human liver microsomes and recombinant human CYP2B6, CYP2C19 and CYP2D6. delta(3)-carene-10-ol 0-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 16415532-0 2005 Localization of rat cytochrome P450 in various tissues and comparison of arachidonic acid metabolism by rat P450 with that by human P450 orthologs. Arachidonic Acid 73-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 16379671-9 2005 Further kinetic analysis revealed that CYP2B6 exhibited the highest activity for Delta(3)-carene 10-hydroxylation. delta-3-carene 81-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 16379671-10 2005 Michaelis-Menten K(m) and V(max) for oxidation of Delta(3)-carene were 0.6 mM and 28.4 nmol/min/nmol P450 using human CYP2B6. delta-3-carene 50-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 16415532-1 2005 Metabolites of arachidonic acid produced by P450 are interesting substances with prominent physiological functions. Arachidonic Acid 15-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 16049129-0 2005 In vitro metabolism of the calmodulin antagonist DY-9760e (3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate) by human liver microsomes: involvement of cytochromes p450 in atypical kinetics and potential drug interactions. 3-(2-(4-(3-chloro-2-methylphenyl)1-piperazinyl)ethyl)5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazol dihydrochloride 3.5 hydrate 49-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 246-250 16267764-1 2005 BACKGROUND: Efavirenz and nelfinavir are metabolized by cytochrome P-450 (CYP) 2B6 and CYP2C19, respectively, with some involvement by CYP3A. efavirenz 12-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-82 16267764-1 2005 BACKGROUND: Efavirenz and nelfinavir are metabolized by cytochrome P-450 (CYP) 2B6 and CYP2C19, respectively, with some involvement by CYP3A. Nelfinavir 26-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-82 16267764-9 2005 Plasma exposure to efavirenz and nelfinavir in each population was significantly associated with the polymorphisms CYP2B6 516G-->T and CYP2C19 681G-->A, respectively. efavirenz 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 16267764-9 2005 Plasma exposure to efavirenz and nelfinavir in each population was significantly associated with the polymorphisms CYP2B6 516G-->T and CYP2C19 681G-->A, respectively. Nelfinavir 33-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 16277416-0 2005 Phytoremediation of metolachlor by transgenic rice plants expressing human CYP2B6. metolachlor 20-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 16277416-3 2005 In particular, CYP2B6 rice plants grown in soil showed tolerance to the chloroacetanilide herbicides alachlor and metolachlor. 2-chloroacetanilide 72-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 16277416-3 2005 In particular, CYP2B6 rice plants grown in soil showed tolerance to the chloroacetanilide herbicides alachlor and metolachlor. alachlor 101-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 16277416-3 2005 In particular, CYP2B6 rice plants grown in soil showed tolerance to the chloroacetanilide herbicides alachlor and metolachlor. metolachlor 114-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 16277416-4 2005 We evaluated the degradation of metolachlor by CYP2B6 rice plants to confirm the metabolic activity of the introduced CYP2B6. metolachlor 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 16277416-4 2005 We evaluated the degradation of metolachlor by CYP2B6 rice plants to confirm the metabolic activity of the introduced CYP2B6. metolachlor 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 16277416-6 2005 In a greenhouse, the ability of CYP2B6 rice plants to remove metolachlor was confirmed in large-scale experiments, in which these plants appeared able to decrease residual quantities of metolachlor in water and soil. metolachlor 61-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 16277416-6 2005 In a greenhouse, the ability of CYP2B6 rice plants to remove metolachlor was confirmed in large-scale experiments, in which these plants appeared able to decrease residual quantities of metolachlor in water and soil. metolachlor 186-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 16368442-12 2005 Hydroxybupropion, the primary active metabolite (t(1/2), approximately 20 hours), is formed by cytochrome P450 (CYP) 2B6. hydroxybupropion 0-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-120 16103134-0 2005 Relative contributions of the five major human cytochromes P450, 1A2, 2C9, 2C19, 2D6, and 3A4, to the hepatic metabolism of the proteasome inhibitor bortezomib. Bortezomib 149-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-63 16103134-3 2005 The relative contributions of the five major human cytochromes P450 (P450s), 1A2, 2C9, 2C19, 2D6, and 3A4 (the focus of the present study), to the metabolism of bortezomib are an important aspect of potential drug interactions. Bortezomib 161-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 16103134-3 2005 The relative contributions of the five major human cytochromes P450 (P450s), 1A2, 2C9, 2C19, 2D6, and 3A4 (the focus of the present study), to the metabolism of bortezomib are an important aspect of potential drug interactions. Bortezomib 161-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-74 15856231-0 2005 P450 induction alters paclitaxel pharmacokinetics and tissue distribution with multiple dosing. Paclitaxel 22-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 16103134-4 2005 Relative activity factor (RAF), chemical inhibition, and immunoinhibition using monoclonal antibodies were three approaches employed to determine the relative contributions of the major human P450s to the net hepatic metabolism of bortezomib. Bortezomib 231-241 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-197 16103134-5 2005 RAFs for the P450 isoform-selective substrates were determined; the ratio of the rate of metabolism of bortezomib with cDNA-expressed P450s versus rate of metabolism with human liver microsomes was normalized with respect to the RAF for each P450 isoform to determine the percentage contributions of the P450s to the net hepatic metabolism of bortezomib. Bortezomib 103-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 16103134-5 2005 RAFs for the P450 isoform-selective substrates were determined; the ratio of the rate of metabolism of bortezomib with cDNA-expressed P450s versus rate of metabolism with human liver microsomes was normalized with respect to the RAF for each P450 isoform to determine the percentage contributions of the P450s to the net hepatic metabolism of bortezomib. Bortezomib 103-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-138 16103134-7 2005 Chemical inhibition and immunoinhibition confirmed that CYP3A4 and CYP2C19 were the major P450s responsible for the hepatic metabolism of bortezomib. Bortezomib 138-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-95 15828049-12 2005 In Supersomes, CYP1A2 was even more efficient in oxidizing o-anisidine than CYP2E1, followed by CYP2B6, 1A1, 2A6, 2D6 and 3A4. 2-anisidine 59-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 16141545-4 2005 The metabolism of clotiazepam was catalyzed by CYP2B6, CYP3A4, CYP2C18, and CYP2C19, and imipramine was metabolized by CYP2D6 most efficiently. clotiazepam 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 15987777-0 2005 Oxidation of tamoxifen by human flavin-containing monooxygenase (FMO) 1 and FMO3 to tamoxifen-N-oxide and its novel reduction back to tamoxifen by human cytochromes P450 and hemoglobin. Tamoxifen 13-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-184 15987777-0 2005 Oxidation of tamoxifen by human flavin-containing monooxygenase (FMO) 1 and FMO3 to tamoxifen-N-oxide and its novel reduction back to tamoxifen by human cytochromes P450 and hemoglobin. tamoxifen N-oxide 84-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-184 15987777-0 2005 Oxidation of tamoxifen by human flavin-containing monooxygenase (FMO) 1 and FMO3 to tamoxifen-N-oxide and its novel reduction back to tamoxifen by human cytochromes P450 and hemoglobin. Tamoxifen 84-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-184 16011913-2 2005 As it is metabolised to hydroxybupropion specifically by CYP2B6, bupropion has also been used as a probe to assess CYP2B6 activity. hydroxybupropion 24-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 16011913-2 2005 As it is metabolised to hydroxybupropion specifically by CYP2B6, bupropion has also been used as a probe to assess CYP2B6 activity. Bupropion 31-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 16011913-9 2005 This assay is more sensitive than currently published methods using HPLC with UV detection for the simultaneous quantitation of bupropion and metabolites and can be used for assessing CYP2B6 activity in vivo following a single dose of bupropion. Bupropion 128-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-190 16011913-9 2005 This assay is more sensitive than currently published methods using HPLC with UV detection for the simultaneous quantitation of bupropion and metabolites and can be used for assessing CYP2B6 activity in vivo following a single dose of bupropion. Bupropion 235-244 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 184-190 15919850-6 2005 CYP2B cells and cDNA-expressed CYP2B enzymes metabolized CPA almost exclusively by 4-hydroxylation, whereas R-IFA and S-IFA were substantially converted to inactive, N-dechloroethylated metabolites. Cyclophosphamide 57-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 15919850-6 2005 CYP2B cells and cDNA-expressed CYP2B enzymes metabolized CPA almost exclusively by 4-hydroxylation, whereas R-IFA and S-IFA were substantially converted to inactive, N-dechloroethylated metabolites. Cyclophosphamide 57-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-36 15980100-9 2005 Although high concentrations of the substrate lauric acid increased BiFC for both P450 2E1 and P450 2C2 with P450 reductase, the concentration dependence did not correlate with reported K(m) values. lauric acid 46-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 15980100-9 2005 Although high concentrations of the substrate lauric acid increased BiFC for both P450 2E1 and P450 2C2 with P450 reductase, the concentration dependence did not correlate with reported K(m) values. lauric acid 46-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 15980100-9 2005 Although high concentrations of the substrate lauric acid increased BiFC for both P450 2E1 and P450 2C2 with P450 reductase, the concentration dependence did not correlate with reported K(m) values. lauric acid 46-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 15856231-2 2005 Studies on the metabolism and disposition of paclitaxel have shown that it is primarily eliminated via hepatic metabolism by P450 enzymes (2C8 and 3A4) to essentially inactive metabolites, and that biliary and gut transport by P-glycoprotein (PGP) as well as urinary elimination of the parent compound play relatively minor roles. Paclitaxel 45-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-129 15856231-8 2005 The effect of paclitaxel treatment on hepatic expression of PGP and P450 isoforms (CYP2C and CYP3A) was determined to elucidate the mechanism by which paclitaxel disposition is altered by previous drug exposure. Paclitaxel 14-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 15856231-8 2005 The effect of paclitaxel treatment on hepatic expression of PGP and P450 isoforms (CYP2C and CYP3A) was determined to elucidate the mechanism by which paclitaxel disposition is altered by previous drug exposure. Paclitaxel 151-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 15983905-1 2005 Although it has not been demonstrated yet, phenytoin is expected to reduce efavirenz exposure through coinduction of cytochrome P450 (CYP) 3A4 and CYP2B6. Phenytoin 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-153 15729289-3 2005 Cytochrome P450 2C11 is an arachidonic acid (AA) epoxygenase expressed in astrocytes, which metabolizes AA to epoxyeicosatrienoic acids (EETs). epoxyeicosatrienoic acids 110-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 15729289-3 2005 Cytochrome P450 2C11 is an arachidonic acid (AA) epoxygenase expressed in astrocytes, which metabolizes AA to epoxyeicosatrienoic acids (EETs). eets 137-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 15919850-0 2005 Enantioselective metabolism and cytotoxicity of R-ifosfamide and S-ifosfamide by tumor cell-expressed cytochromes P450. r-ifosfamide 48-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-118 15919850-0 2005 Enantioselective metabolism and cytotoxicity of R-ifosfamide and S-ifosfamide by tumor cell-expressed cytochromes P450. Ifosfamide 65-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-118 15919850-2 2005 Hepatic cytochrome P450 (P450) enzymes exhibit enantioselective preferences in the metabolism of R-IFA and S-IFA; however, the impact of this selectivity on P450-dependent anticancer activity is not known. r-ifa 97-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 15919850-2 2005 Hepatic cytochrome P450 (P450) enzymes exhibit enantioselective preferences in the metabolism of R-IFA and S-IFA; however, the impact of this selectivity on P450-dependent anticancer activity is not known. r-ifa 97-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 15919850-2 2005 Hepatic cytochrome P450 (P450) enzymes exhibit enantioselective preferences in the metabolism of R-IFA and S-IFA; however, the impact of this selectivity on P450-dependent anticancer activity is not known. r-ifa 97-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 15919850-2 2005 Hepatic cytochrome P450 (P450) enzymes exhibit enantioselective preferences in the metabolism of R-IFA and S-IFA; however, the impact of this selectivity on P450-dependent anticancer activity is not known. Ifosfamide 107-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 15919850-2 2005 Hepatic cytochrome P450 (P450) enzymes exhibit enantioselective preferences in the metabolism of R-IFA and S-IFA; however, the impact of this selectivity on P450-dependent anticancer activity is not known. Ifosfamide 107-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 15919850-2 2005 Hepatic cytochrome P450 (P450) enzymes exhibit enantioselective preferences in the metabolism of R-IFA and S-IFA; however, the impact of this selectivity on P450-dependent anticancer activity is not known. Ifosfamide 107-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 15919850-4 2005 Tumor cells expressing P450 enzyme CYP3A4 were the most sensitive to R-IFA cytotoxicity, whereas tumor cells expressing CYP2B1 or CYP2B6 were most sensitive to cyclophosphamide (CPA), an isomer of IFA. Cyclophosphamide 160-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 15919850-4 2005 Tumor cells expressing P450 enzyme CYP3A4 were the most sensitive to R-IFA cytotoxicity, whereas tumor cells expressing CYP2B1 or CYP2B6 were most sensitive to cyclophosphamide (CPA), an isomer of IFA. Cyclophosphamide 178-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 15919850-4 2005 Tumor cells expressing P450 enzyme CYP3A4 were the most sensitive to R-IFA cytotoxicity, whereas tumor cells expressing CYP2B1 or CYP2B6 were most sensitive to cyclophosphamide (CPA), an isomer of IFA. Cyclophosphamide 178-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 15821045-1 2005 Ifosfamide nephrotoxicity is attributed to the formation of a toxic metabolite, chloroacetaldehyde, via N-dechloroethylation, a reaction that is purportedly catalyzed by CYP3A and CYP2B6. Ifosfamide 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 180-186 16278191-4 2005 Additionally, a good correlation was observed with CYP2C19, CYP2C8 and CYP2B6 at a ketobemidone concentration of 50 microM. ketobemidone 83-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 15821045-1 2005 Ifosfamide nephrotoxicity is attributed to the formation of a toxic metabolite, chloroacetaldehyde, via N-dechloroethylation, a reaction that is purportedly catalyzed by CYP3A and CYP2B6. chloroacetaldehyde 80-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 180-186 15802384-9 2005 Cerivastatin, simvastatin, fluvastatin, and atorvastatin induced CYP2B6 mRNA in stable hCAR-expressed FLC7 cells but not in original FLC7 cells. cerivastatin 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 15802384-9 2005 Cerivastatin, simvastatin, fluvastatin, and atorvastatin induced CYP2B6 mRNA in stable hCAR-expressed FLC7 cells but not in original FLC7 cells. Simvastatin 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 15802384-9 2005 Cerivastatin, simvastatin, fluvastatin, and atorvastatin induced CYP2B6 mRNA in stable hCAR-expressed FLC7 cells but not in original FLC7 cells. Fluvastatin 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 15802384-9 2005 Cerivastatin, simvastatin, fluvastatin, and atorvastatin induced CYP2B6 mRNA in stable hCAR-expressed FLC7 cells but not in original FLC7 cells. Atorvastatin 44-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 15821045-1 2005 Ifosfamide nephrotoxicity is attributed to the formation of a toxic metabolite, chloroacetaldehyde, via N-dechloroethylation, a reaction that is purportedly catalyzed by CYP3A and CYP2B6. Nitrogen 104-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 180-186 15834119-3 2005 In this assay, the P450 is incubated with a fixed amount of radiolabeled cholesterol and varying concentrations of cold cholesterol followed by separation of free and protein-bound cholesterol via filtration through a membrane. Cholesterol 73-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 15712360-0 2005 A cytochrome P450 2B6 meditated gene therapy strategy to enhance the effects of radiation or cyclophosphamide when combined with the bioreductive drug AQ4N. Cyclophosphamide 93-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 2-21 15712360-4 2005 We have therefore examined the potential of a CYP2B6-mediated GDEPT strategy to enhance the anti-tumour effect of the combination of AQ4N with radiation or cyclophosphamide (CPA). AQ4N 133-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 15712360-4 2005 We have therefore examined the potential of a CYP2B6-mediated GDEPT strategy to enhance the anti-tumour effect of the combination of AQ4N with radiation or cyclophosphamide (CPA). Cyclophosphamide 156-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 15712360-4 2005 We have therefore examined the potential of a CYP2B6-mediated GDEPT strategy to enhance the anti-tumour effect of the combination of AQ4N with radiation or cyclophosphamide (CPA). Cyclophosphamide 174-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 15712360-8 2005 RESULTS: When CYP2B6 was transfected into RIF-1 cells and treated with AQ4N under hypoxic conditions there was a significant increase in DNA damage (measured by the ACA) compared with non-transfected cells. AQ4N 71-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 15802384-1 2005 Constitutive active (or androstane) receptor (CAR, NR1I3), a member of the nuclear receptor family, is a major regulator for induction of cytochrome P450 2B (CYP2B) genes by phenobarbital. Phenobarbital 174-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-156 15802384-1 2005 Constitutive active (or androstane) receptor (CAR, NR1I3), a member of the nuclear receptor family, is a major regulator for induction of cytochrome P450 2B (CYP2B) genes by phenobarbital. Phenobarbital 174-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-163 15712360-9 2005 In vivo, a single intra-tumoural injection of a CYP2B6 vector construct significantly enhanced tumour growth delay in combination with AQ4N (100 mg/kg) and 10 Gy X-rays. AQ4N 135-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-54 15834119-3 2005 In this assay, the P450 is incubated with a fixed amount of radiolabeled cholesterol and varying concentrations of cold cholesterol followed by separation of free and protein-bound cholesterol via filtration through a membrane. Cholesterol 120-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 15712360-10 2005 AQ4N (100 mg/kg) and CPA (100 mg/kg) with CYP2B6 and CYPRED also enhanced tumour growth delay; this effect became significant when the schedule was repeated 14 days later (p = 0.0197). AQ4N 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 15834119-3 2005 In this assay, the P450 is incubated with a fixed amount of radiolabeled cholesterol and varying concentrations of cold cholesterol followed by separation of free and protein-bound cholesterol via filtration through a membrane. Cholesterol 120-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 15712360-10 2005 AQ4N (100 mg/kg) and CPA (100 mg/kg) with CYP2B6 and CYPRED also enhanced tumour growth delay; this effect became significant when the schedule was repeated 14 days later (p = 0.0197). Cyclophosphamide 21-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 15761118-6 2005 By combining in vitro transfection methods and quantitative analyses of gene expression in cell lines and primary human hepatocytes, we here show that artemisinin drugs activate human PXR as well as human and mouse CAR and induce the expression of CYP2B6, CYP3A4, and MDR1 in primary human hepatocytes and in the human intestinal cell line LS174T. artemisinin 151-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 248-254 15961986-0 2005 Effect of clopidogrel and ticlopidine on cytochrome P450 2B6 activity as measured by bupropion hydroxylation. Clopidogrel 10-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-60 15961986-0 2005 Effect of clopidogrel and ticlopidine on cytochrome P450 2B6 activity as measured by bupropion hydroxylation. Ticlopidine 26-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-60 15961986-0 2005 Effect of clopidogrel and ticlopidine on cytochrome P450 2B6 activity as measured by bupropion hydroxylation. Bupropion 85-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-60 15961986-1 2005 OBJECTIVE: Our objective was to study the effect of the antiplatelet agents clopidogrel and ticlopidine on bupropion (INN, amfebutamone) hydroxylation, a probe reaction for cytochrome P450 (CYP) 2B6 activity. Clopidogrel 76-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-198 15961986-1 2005 OBJECTIVE: Our objective was to study the effect of the antiplatelet agents clopidogrel and ticlopidine on bupropion (INN, amfebutamone) hydroxylation, a probe reaction for cytochrome P450 (CYP) 2B6 activity. Ticlopidine 92-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-198 15961986-1 2005 OBJECTIVE: Our objective was to study the effect of the antiplatelet agents clopidogrel and ticlopidine on bupropion (INN, amfebutamone) hydroxylation, a probe reaction for cytochrome P450 (CYP) 2B6 activity. Bupropion 107-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-198 15961986-4 2005 Plasma concentrations of bupropion and its CYP2B6-catalyzed metabolite, hydroxybupropion, were measured for up to 72 hours. hydroxybupropion 72-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 15961986-8 2005 CONCLUSIONS: Both clopidogrel and ticlopidine significantly inhibited the CYP2B6-catalyzed bupropion hydroxylation. Clopidogrel 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 15961986-8 2005 CONCLUSIONS: Both clopidogrel and ticlopidine significantly inhibited the CYP2B6-catalyzed bupropion hydroxylation. Ticlopidine 34-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 15961986-8 2005 CONCLUSIONS: Both clopidogrel and ticlopidine significantly inhibited the CYP2B6-catalyzed bupropion hydroxylation. Bupropion 91-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 15961986-9 2005 Patients receiving either clopidogrel or ticlopidine are likely to require dose adjustments when treated with drugs primarily metabolized by CYP2B6. Clopidogrel 26-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 15961986-9 2005 Patients receiving either clopidogrel or ticlopidine are likely to require dose adjustments when treated with drugs primarily metabolized by CYP2B6. Ticlopidine 41-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 15774560-6 2005 Type II P450 enzymes, found in the endoplasmic reticulum, receive electrons from NADPH via P450 oxidoreductase (POR), which contains two flavin moieties. NADP 81-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-12 15774560-6 2005 Type II P450 enzymes, found in the endoplasmic reticulum, receive electrons from NADPH via P450 oxidoreductase (POR), which contains two flavin moieties. 4,6-dinitro-o-cresol 137-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-12 15853388-3 2005 At germination, R(1) seeds of transgenic rice plants expressing CYP2B6 (CYP2B6 rice) showed a high tolerance to 5 microM metolachlor, a preemergence herbicide that is degraded by CYP2B6. metolachlor 121-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 15853388-4 2005 Thin-layer chromatography after culture with (14)C-labeled metolachlor revealed that the amounts of residual metolachlor decreased in plant tissues and the medium of CYP2B6 rice faster than those of untransformed Nipponbare. metolachlor 109-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 15853388-3 2005 At germination, R(1) seeds of transgenic rice plants expressing CYP2B6 (CYP2B6 rice) showed a high tolerance to 5 microM metolachlor, a preemergence herbicide that is degraded by CYP2B6. metolachlor 121-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 15853388-5 2005 CYP2B6 rice plants were able to grow in the presence of 13 out of 17 herbicides: five chloroacetamides and mefenacet, pyributicarb, amiprofos-methyl, trifluralin, pendimethalin, norflurazon, and chlorotoluron. chloroacetamide 86-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15853388-5 2005 CYP2B6 rice plants were able to grow in the presence of 13 out of 17 herbicides: five chloroacetamides and mefenacet, pyributicarb, amiprofos-methyl, trifluralin, pendimethalin, norflurazon, and chlorotoluron. mefenacet 107-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15853388-3 2005 At germination, R(1) seeds of transgenic rice plants expressing CYP2B6 (CYP2B6 rice) showed a high tolerance to 5 microM metolachlor, a preemergence herbicide that is degraded by CYP2B6. metolachlor 121-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 15853388-5 2005 CYP2B6 rice plants were able to grow in the presence of 13 out of 17 herbicides: five chloroacetamides and mefenacet, pyributicarb, amiprofos-methyl, trifluralin, pendimethalin, norflurazon, and chlorotoluron. Pyributicarb 118-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15853388-5 2005 CYP2B6 rice plants were able to grow in the presence of 13 out of 17 herbicides: five chloroacetamides and mefenacet, pyributicarb, amiprofos-methyl, trifluralin, pendimethalin, norflurazon, and chlorotoluron. amiprophos methyl 132-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15853388-4 2005 Thin-layer chromatography after culture with (14)C-labeled metolachlor revealed that the amounts of residual metolachlor decreased in plant tissues and the medium of CYP2B6 rice faster than those of untransformed Nipponbare. metolachlor 59-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 15778010-3 2005 Using Escherichia coli-expressed human P450, our results demonstrated that safrole was a non-selective inhibitor of CYP1A2, CYP2A6, CYP2D6, CYP2E1, and CYP3A4 in the IC(50) order CYP2E1 < CYP1A2 < CYP2A6 < CYP3A4 < CYP2D6. Safrole 75-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 15853388-5 2005 CYP2B6 rice plants were able to grow in the presence of 13 out of 17 herbicides: five chloroacetamides and mefenacet, pyributicarb, amiprofos-methyl, trifluralin, pendimethalin, norflurazon, and chlorotoluron. Trifluralin 150-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15853388-5 2005 CYP2B6 rice plants were able to grow in the presence of 13 out of 17 herbicides: five chloroacetamides and mefenacet, pyributicarb, amiprofos-methyl, trifluralin, pendimethalin, norflurazon, and chlorotoluron. pendimethalin 163-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15853388-5 2005 CYP2B6 rice plants were able to grow in the presence of 13 out of 17 herbicides: five chloroacetamides and mefenacet, pyributicarb, amiprofos-methyl, trifluralin, pendimethalin, norflurazon, and chlorotoluron. norflurazone 178-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15853388-5 2005 CYP2B6 rice plants were able to grow in the presence of 13 out of 17 herbicides: five chloroacetamides and mefenacet, pyributicarb, amiprofos-methyl, trifluralin, pendimethalin, norflurazon, and chlorotoluron. chlortoluron 195-208 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15743975-9 2005 Among the 13 P450 isoforms tested, CYP 3A4, 2C8, 3A5, and 3A7 produced Nor-BUP. norbuprenorphine 71-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 15746946-2 2005 Our approach is a P450-based VDEPT that consists of using cyclophosphamide (CPA) as a prodrug and a Cytochrome P450 2B6/NADPH cytochrome P450 reductase fusion protein (CYP2B6/RED) as a prodrug-activating enzyme. Cyclophosphamide 76-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 168-174 15746946-5 2005 The CYP2B6/RED fusion protein was detected by Western blot, its mRNA by Northern blot, and its heme incorporation into an active form by spectral analysis. Heme 95-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 15746946-7 2005 After infection and treatment with CPA, in cell lines with low endogenous RED, the fusion protein mediated significantly higher CPA-induced cytotoxicity compared to cells expressing solely CYP2B6. Cyclophosphamide 35-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-195 15616152-0 2005 Metabolism of melatonin by human cytochromes p450. Melatonin 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-49 16012075-8 2005 Phenobarbital-mediated responsiveness of cytochrome P450 2B6, a potential indicator of hepatocyte differentiation status, was markedly higher in overlaid relative to non-overlaid hepatocytes. Phenobarbital 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-60 15703377-5 2005 To further validate S. cerevisiae as a model for exploring mammalian P450 turnover, the degradation of phenobarbital-inducible liver CYP2B1, an enzyme reportedly degraded via the rat hepatic autophagic-lysosomal pathway, was examined in a yeast strain (pep4delta) deficient in vacuolar degradation and its isogenic wild-type control (PEP4). Phenobarbital 103-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-73 15819722-0 2005 StAR and progesterone producing enzymes (3beta-hydroxysteroid dehydrogenase and cholesterol side-chain cleavage cytochromes P450) in human epithelial ovarian carcinoma: immunohistochemical and real-time PCR studies. Cholesterol 80-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 15616152-9 2005 These findings confirm the suggestion of others that CYP1A2 is probably the foremost hepatic P450 in the 6-hydroxylation of MEL and a single report that CYP1A1 is also able to mediate this reaction. Melatonin 124-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 15787531-0 2005 Cyclopropyl containing fatty acids as mechanistic probes for cytochromes P450. cyclopropyl containing fatty acids 0-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 15752707-1 2005 To probe whether the nature of the substrate can directly influence the spectral properties of oxyferrous cytochrome P450-CAM, the complex has been investigated in the absence and in the presence of the natural substrate (1R)-camphor (camphor) and of several camphor analogs. Camphor 226-233 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-121 15752707-4 2005 The MCD spectrum of camphor-bound oxyferrous P450-CAM is similar to that of the substrate-free oxyferrous enzyme, but the spectrum of the oxyferrous enzyme differs detectably in the presence of substrate analogs. Camphor 20-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-49 15752707-7 2005 The present results indicate that the structures of the camphor analogs can sensitively influence the physical (spectroscopic) properties of the P450 dioxygen complex and could also affect its reactivity. Camphor 56-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-149 15752707-7 2005 The present results indicate that the structures of the camphor analogs can sensitively influence the physical (spectroscopic) properties of the P450 dioxygen complex and could also affect its reactivity. Oxygen 150-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-149 15787531-6 2005 Oxidation of these probes by P450(BM3) and P450(BioI) gives results consistent with a radical but not a cationic intermediate in fatty acid hydroxylation by these enzymes. Fatty Acids 129-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 15787531-6 2005 Oxidation of these probes by P450(BM3) and P450(BioI) gives results consistent with a radical but not a cationic intermediate in fatty acid hydroxylation by these enzymes. Fatty Acids 129-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-53 15651036-8 2005 The modified scoring functions will be useful in docking applications on P450 enzymes and other heme binding proteins. Heme 96-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 15755146-3 2005 In the biosynthesis of the fungal carcinogen, aflatoxin, six cytochromes P450 are encoded by the biosynthetic gene cluster. Aflatoxins 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 15686361-0 2005 P(450)/NADPH/O(2)- and P(450)/PhIO-catalyzed N-dealkylations are mechanistically distinct. Nitrogen 7-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15572372-0 2005 AMP-activated protein kinase mediates phenobarbital induction of CYP2B gene expression in hepatocytes and a newly derived human hepatoma cell line. Phenobarbital 38-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-70 15572372-8 2005 In this report, we show that addition of 5-amino-imidazole carboxamide riboside, an AMPK activator, to WGA and human hepatocytes induces CYP2B6 gene expression. 5-amino-imidazole carboxamide riboside 41-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 15572372-9 2005 Expression of a constitutively active form of AMPK mimics the PB induction of CYP2B6 and CYP2B1 gene expression. Phenobarbital 62-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 15572372-12 2005 Our data strongly support a role for AMPK in the PB induction of CYP2B gene expression and provide new insights into the regulation of gene expression by barbiturate drugs. Phenobarbital 49-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-70 15686361-5 2005 Herein we present the first experimental evidence to demonstrate that the NADPH/O2- and PhIO-supported P450 N-dealkylations are mechanistically distinct and, thus, the P450/PhIO system may not be a good mechanistic model for P450/NADPH/O2-catalyzed N-dealkylations. Oxygen 80-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 225-235 15563456-1 2005 The constitutive active receptor (CAR) regulates the induction of the cytochrome P450 2B6 (CYP2B6) gene by phenobarbital-type inducers, such as 1,4 bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) via the distal phenobarbital-responsive enhancer module (PBREM, at -1732/-1685 bp). Phenobarbital 107-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-89 15563456-1 2005 The constitutive active receptor (CAR) regulates the induction of the cytochrome P450 2B6 (CYP2B6) gene by phenobarbital-type inducers, such as 1,4 bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) via the distal phenobarbital-responsive enhancer module (PBREM, at -1732/-1685 bp). Phenobarbital 107-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 15563456-1 2005 The constitutive active receptor (CAR) regulates the induction of the cytochrome P450 2B6 (CYP2B6) gene by phenobarbital-type inducers, such as 1,4 bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) via the distal phenobarbital-responsive enhancer module (PBREM, at -1732/-1685 bp). 1,4-bis(2-(3,5-dichloropyridyloxy))benzene 144-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-89 15563456-1 2005 The constitutive active receptor (CAR) regulates the induction of the cytochrome P450 2B6 (CYP2B6) gene by phenobarbital-type inducers, such as 1,4 bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) via the distal phenobarbital-responsive enhancer module (PBREM, at -1732/-1685 bp). 1,4-bis(2-(3,5-dichloropyridyloxy))benzene 144-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 15563456-1 2005 The constitutive active receptor (CAR) regulates the induction of the cytochrome P450 2B6 (CYP2B6) gene by phenobarbital-type inducers, such as 1,4 bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) via the distal phenobarbital-responsive enhancer module (PBREM, at -1732/-1685 bp). 1,4-bis(2-(3,5-dichloropyridyloxy))benzene 188-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-89 15563456-1 2005 The constitutive active receptor (CAR) regulates the induction of the cytochrome P450 2B6 (CYP2B6) gene by phenobarbital-type inducers, such as 1,4 bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) via the distal phenobarbital-responsive enhancer module (PBREM, at -1732/-1685 bp). 1,4-bis(2-(3,5-dichloropyridyloxy))benzene 188-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 15563456-1 2005 The constitutive active receptor (CAR) regulates the induction of the cytochrome P450 2B6 (CYP2B6) gene by phenobarbital-type inducers, such as 1,4 bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) via the distal phenobarbital-responsive enhancer module (PBREM, at -1732/-1685 bp). Phenobarbital 212-225 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-89 15563456-1 2005 The constitutive active receptor (CAR) regulates the induction of the cytochrome P450 2B6 (CYP2B6) gene by phenobarbital-type inducers, such as 1,4 bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) via the distal phenobarbital-responsive enhancer module (PBREM, at -1732/-1685 bp). Phenobarbital 212-225 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 15547048-3 2005 Using P450 isoform-selective inhibitors and recombinant heterologously expressed enzymes, it was demonstrated that several P450 enzymes catalyzed sertraline N-demethylation, with CYP2B6 contributing the greatest extent, and lesser contributions from CYP2C19, CYP2C9, CYP3A4, and CYP2D6. sertraline n 146-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 179-185 15652242-0 2005 Inhibition of human CYP2B6 by N,N",N""-triethylenethiophosphoramide is irreversible and mechanism-based. Thiotepa 30-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 15652242-2 2005 Previous studies demonstrated partial inhibition by thioTEPA of the cytochrome P4502B6 (CYP2B6)-catalyzed 4-hydroxylation of cyclophosphamide, which is required for its bioactivation. Thiotepa 52-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 15652242-2 2005 Previous studies demonstrated partial inhibition by thioTEPA of the cytochrome P4502B6 (CYP2B6)-catalyzed 4-hydroxylation of cyclophosphamide, which is required for its bioactivation. Cyclophosphamide 125-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 15652242-3 2005 The aim of our study was to investigate the detailed mechanism of CYP2B6 inhibition by thioTEPA. Thiotepa 87-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 15652242-4 2005 Using human liver microsomes and recombinant P450 enzymes we confirmed potent inhibition of CYP2B6 enzyme activity determined with bupropion as substrate. Bupropion 131-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 15652242-5 2005 ThioTEPA was found to inhibit CYP2B6 activity in a time- and concentration-dependent manner. Thiotepa 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 15652242-6 2005 The loss of CYP2B6 activity was NADPH-dependent and could not be restored by extensive dialysis. NADP 32-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 15652242-10 2005 Inactivated CYP2B6 did not lose its ability to form a CO-reduced complex suggesting a modification of the apoprotein, which is common for sulfur-containing compounds. Sulfur 138-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 15770070-1 2005 Cytochrome P450 (P450) constitutes a superfamily of enzymes which activate dioxygen and carry out monooxygenation reactions of large numbers of endogenous and xenobiotic compounds. Oxygen 75-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 15822184-2 2005 In the course of these reactions, the enzymes generate reactive O(2) species and/or reactive metabolic products that can attack the P450 heme and/or protein moiety and structurally and functionally damage the enzyme. reactive o(2) species 55-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-136 15822184-2 2005 In the course of these reactions, the enzymes generate reactive O(2) species and/or reactive metabolic products that can attack the P450 heme and/or protein moiety and structurally and functionally damage the enzyme. Heme 137-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-136 23045123-1 2005 NADH cytochrome b(5) reductase (b(5)R; EC 1.6.2.2; Diaphorase I; NADH: ferricytochrome b(5) oxidoreductase) is an FAD-containing protein, which, along with the hemoprotein cytochrome b(5) (cyt b(5)), mediates electron transfer from NADH to fatty acid desaturases, P450 oxidases, methemoglobin, and ascorbyl free radical. NAD 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 264-268 23045123-1 2005 NADH cytochrome b(5) reductase (b(5)R; EC 1.6.2.2; Diaphorase I; NADH: ferricytochrome b(5) oxidoreductase) is an FAD-containing protein, which, along with the hemoprotein cytochrome b(5) (cyt b(5)), mediates electron transfer from NADH to fatty acid desaturases, P450 oxidases, methemoglobin, and ascorbyl free radical. DL-Ascorbic acid 298-306 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 264-268 23045123-1 2005 NADH cytochrome b(5) reductase (b(5)R; EC 1.6.2.2; Diaphorase I; NADH: ferricytochrome b(5) oxidoreductase) is an FAD-containing protein, which, along with the hemoprotein cytochrome b(5) (cyt b(5)), mediates electron transfer from NADH to fatty acid desaturases, P450 oxidases, methemoglobin, and ascorbyl free radical. radical 312-319 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 264-268 15486075-1 2005 Previous studies in our laboratory showed that among cDNA-expressed human cytochrome P450 (P450) supersomes, CYP2C19 was the most active in methoxychlor-O-demethylation. methoxychlor-o 140-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-96 15604546-4 2005 The second case with mild acne had a soaring serum level of total testosterone >9,000 ng/dl derived from an androgen-secreting adrenal adenoma overexpressing steroidogenic acute regulatory protein, P450 side-chain cleavage enzyme and aromatase. Testosterone 66-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 201-205 16010857-19 2005 To get an overview on the postnatal development of the most important phase I and phase II reactions and also for the heme biosynthetic pathway as a prerequisite for the P450 synthesis, a compilation of literature data similar to a score was constructed with the aim to recognize parallel developments, possible common control and regulation mechanisms, cp. Heme 118-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-174 21676740-8 2005 We propose hypotheses suggesting that nitrate could alter steroidogenesis by 1) conversion to nitrite and nitric oxide in the mitochondria, the site of initial steroid synthesis, 2) altering Cl(-) ion concentrations in the cell by substituting for Cl(-) in the membrane transport pump or 3) binding to the heme region of various P450 enzymes associated with steroidogenesis and altering enzymatic action. Nitrates 38-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 329-333 15656694-10 2005 Interactions with the alkylating agents cyclophosphamide and ifosfamide are complicated as a result of the involvement of the CYP3A4 and CYP2B6 isoenzymes in both the metabolic activation of these drugs and the generation of potentially neurotoxic metabolites. Cyclophosphamide 40-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 15656694-10 2005 Interactions with the alkylating agents cyclophosphamide and ifosfamide are complicated as a result of the involvement of the CYP3A4 and CYP2B6 isoenzymes in both the metabolic activation of these drugs and the generation of potentially neurotoxic metabolites. Ifosfamide 61-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 20021072-7 2005 In contrast, CYP2B6, the human orthologue of the rodent phenobarbital-inducible P450 2B, is known to be inducible by a range of substances, but our recent studies also show a high degree of genetic polymorphism. Phenobarbital 56-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 15864119-1 2005 BACKGROUND: Efavirenz (EFV) and nevirapine (NVP) are metabolized by cytochrome P450 2B6 (CYP2B6). Nevirapine 32-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-87 15864119-1 2005 BACKGROUND: Efavirenz (EFV) and nevirapine (NVP) are metabolized by cytochrome P450 2B6 (CYP2B6). Nevirapine 32-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-95 15864119-6 2005 RESULTS AND CONCLUSIONS: CYP2B6 516TT was associated with greater plasma and intracellular exposure to EFV, and greater plasma exposure to NVP. efavirenz 103-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 15849716-6 2005 Pharmacodynamic modeling revealed EC50 values statistically significant between UGT1A1 and CYP2B6 after treatment with PB, but not statistically distinguishable between UGT1A1 and CYP"s 1A2 or 3A4 after treatment with 3-methylchloranthrene or rifampicin, respectively. Lead 119-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 15864119-1 2005 BACKGROUND: Efavirenz (EFV) and nevirapine (NVP) are metabolized by cytochrome P450 2B6 (CYP2B6). efavirenz 23-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-87 15864119-1 2005 BACKGROUND: Efavirenz (EFV) and nevirapine (NVP) are metabolized by cytochrome P450 2B6 (CYP2B6). efavirenz 23-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-95 15622315-9 2004 The CYP2B6 T/T genotype at position 516 (GlnHis) was more common in African-Americans (20%) than in European-Americans (3%), and was associated with greater efavirenz plasma exposure (P < 0.0001). glnhis 41-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 15548381-8 2004 These results suggest that CYP3A4 and CYP2B6 regulation through PXR activation by persistent pesticides may have an impact on the metabolism of xenobiotic agents and endogenous steroid hormones. Steroids 177-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-44 15365656-9 2004 CONCLUSION: CYP2B6, CYP2C8, CYP2D6, and CYP3A4 catalyze LOP N-demethylation in human liver microsomes, and among them, CYP2C8 and CYP3A4 may play a crucial role in LOP metabolism at the therapeutic concentrations of LOP. Nitrogen 2-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 15632987-0 2004 Hydrogen bonding in human p450-substrate interactions: a major contribution to binding affinity. Hydrogen 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 15632987-1 2004 The importance of hydrogen bonding, a relatively strong intermolecular force of attraction between molecules in biological systems, is discussed in the respect of P450 substrate affinity towards one or more of the human P450 enzymes that are generally associated with drug and other xenobiotic metabolism. Hydrogen 18-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-167 15632987-1 2004 The importance of hydrogen bonding, a relatively strong intermolecular force of attraction between molecules in biological systems, is discussed in the respect of P450 substrate affinity towards one or more of the human P450 enzymes that are generally associated with drug and other xenobiotic metabolism. Hydrogen 18-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 220-224 15632987-3 2004 It is thus possible to estimate the hydrogen bond contribution to P450 enzyme-substrate binding affinity based on modelled interactions and by use of these relatively simple formulae, particularly when employed in conjunction with substrate-lipophilicity relationships. Hydrogen 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 15382119-4 2004 We show that IL-1beta decreases CAR expression and decreases phenobarbital- or bilirubin-mediated induction of CYP2B6, CYP2C9, CYP3A4, UGT1A1, GSTA1, GSTA2, and SLC21A6 messenger RNA. Phenobarbital 61-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 15382119-4 2004 We show that IL-1beta decreases CAR expression and decreases phenobarbital- or bilirubin-mediated induction of CYP2B6, CYP2C9, CYP3A4, UGT1A1, GSTA1, GSTA2, and SLC21A6 messenger RNA. Bilirubin 79-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 15258138-1 2004 Alpha-thujone (1alpha) and beta-thujone (1beta) were used to investigate the mechanism of hydrocarbon hydroxylation by cytochromes P-450(cam) (CYP101) and P-450(BM3) (CYP102). beta-thujone 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-165 15378224-2 2004 METHODS: The inhibitory potency of memantine on CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 activities was examined with specific probe drugs in HLM and recombinant P450s. Memantine 35-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 15378224-4 2004 RESULTS: In HLM, memantine inhibited CYP2B6 and CYP2D6 activities, with KI (IC50) values of 76.7 (279.7) and 94.9 (368.7) microM, respectively. Memantine 17-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 15378224-11 2004 CONCLUSION: Memantine exerts selective inhibition of CYP2B6 activity at clinically relevant concentrations, suggesting the potential for clinically significant drug interactions. Memantine 12-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 15378224-13 2004 Moreover, memantine represents a new, potent, selective inhibitor of recombinant CYP2B6, which may prove useful for screening purposes during early phases of in vitro drug metabolism studies with new chemical entities. Memantine 10-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 15764407-6 2004 Recombinant human CYP2B6 possessed the highest desulphuration activity for chlorpyrifos, whereas CYP2C19 had the highest dearylation activity. Chlorpyrifos 75-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 15364537-8 2004 Primary cultures of hepatocytes were treated with beta-naphthoflavone, an inducer of CYP1A2, phenobarbital or rifampin, both of which induce CYP2B6, CYP2C9, CYP2C19, and CYP3A4, albeit it to different extents. beta-Naphthoflavone 50-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 15764407-7 2004 In contrast, both desulphuration and dearylation of diazinon were catalysed at similar rates, in the rank order CYP2C19 > CYP1A2 > CYP2B6 > CYP3A4. Diazinon 52-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 15364537-8 2004 Primary cultures of hepatocytes were treated with beta-naphthoflavone, an inducer of CYP1A2, phenobarbital or rifampin, both of which induce CYP2B6, CYP2C9, CYP2C19, and CYP3A4, albeit it to different extents. Rifampin 110-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 15364541-9 2004 Nicotine, the psychoactive and addictive chemical in cigarettes, and a known inducer of brain CYP2B6, was an efficacious activator of PXR and inducer of CYP3A4 transcription. Nicotine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 15371986-0 2004 Role of hepatic and intestinal cytochrome P450 3A and 2B6 in the metabolism, disposition, and miotic effects of methadone. Methadone 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-57 15371986-22 2004 In vitro experiments showed a predominant role for both CYP3A4 and CYP2B6 in liver microsomal methadone N -demethylation. Methadone 94-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 15371986-26 2004 Interindividual variability and drug interactions affecting intestinal transporter and hepatic CYP3A and CYP2B6 activity may alter methadone disposition. Methadone 131-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-111 15121764-9 2004 CYP2B6 could only be protected from the tTEPA-dependent inactivation by the 2B6-specific substrate bupropion but not by other substrates of CYP2B such as benzphetamine, testosterone, or 7-ethoxycoumarin. Bupropion 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15319333-0 2004 CYP2B6, CYP3A4, and CYP2C19 are responsible for the in vitro N-demethylation of meperidine in human liver microsomes. Nitrogen 61-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15319333-0 2004 CYP2B6, CYP3A4, and CYP2C19 are responsible for the in vitro N-demethylation of meperidine in human liver microsomes. Meperidine 80-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15319333-4 2004 After normalization by its relative abundance in human liver microsomes, CYP2B6, CYP3A4, and CYP2C19 accounted for 57, 28, and 15% of the total intrinsic clearance of meperidine. Meperidine 167-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 15319333-8 2004 An anti-CYP2B6 antibody inhibited normeperidine formation by 46%. normeperidine 34-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 15319333-10 2004 Experiments with thiotepa and ketoconazole suggested inhibition of microsomal CYP2B6 and CYP3A4 activity, whereas studies with fluvoxamine (a substrate of CYP2C19) were inconclusive due to lack of specificity. Thiotepa 17-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 15319333-10 2004 Experiments with thiotepa and ketoconazole suggested inhibition of microsomal CYP2B6 and CYP3A4 activity, whereas studies with fluvoxamine (a substrate of CYP2C19) were inconclusive due to lack of specificity. Ketoconazole 30-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 15319333-11 2004 We conclude that normeperidine formation in human liver microsomes is mainly catalyzed by CYP2B6 and CYP3A4, with a minor contribution from CYP2C19. normeperidine 17-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 15319341-0 2004 Kinetics and regulation of cytochrome P450-mediated etoposide metabolism. Etoposide 52-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 15319341-2 2004 The cytochrome P450 (P450)-derived catechol and quinone metabolites of etoposide may be important in the damage to the MLL (mixed lineage leukemia) gene and other genes resulting in leukemia-associated chromosomal translocations. catechol 35-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 15319341-2 2004 The cytochrome P450 (P450)-derived catechol and quinone metabolites of etoposide may be important in the damage to the MLL (mixed lineage leukemia) gene and other genes resulting in leukemia-associated chromosomal translocations. catechol 35-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 15319341-2 2004 The cytochrome P450 (P450)-derived catechol and quinone metabolites of etoposide may be important in the damage to the MLL (mixed lineage leukemia) gene and other genes resulting in leukemia-associated chromosomal translocations. quinone 48-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 15319341-2 2004 The cytochrome P450 (P450)-derived catechol and quinone metabolites of etoposide may be important in the damage to the MLL (mixed lineage leukemia) gene and other genes resulting in leukemia-associated chromosomal translocations. quinone 48-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 15319341-2 2004 The cytochrome P450 (P450)-derived catechol and quinone metabolites of etoposide may be important in the damage to the MLL (mixed lineage leukemia) gene and other genes resulting in leukemia-associated chromosomal translocations. Etoposide 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 15319341-2 2004 The cytochrome P450 (P450)-derived catechol and quinone metabolites of etoposide may be important in the damage to the MLL (mixed lineage leukemia) gene and other genes resulting in leukemia-associated chromosomal translocations. Etoposide 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 15319341-4 2004 CYP3A4 was found to play a major role in etoposide metabolism (K(m) = 77.7 +/- 27.8 microM; V(max) = 314 +/- 84 pmol of catechol/min/nmol of P450). Etoposide 41-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 15319341-6 2004 9 +/- 3.1 microM; V(max) = 19.4 +/- 0.4 pmol of catechol/min/nmol of P450) may be involved in etoposide metabolism at therapeutic concentrations of free drug. catechol 48-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-73 15319341-6 2004 9 +/- 3.1 microM; V(max) = 19.4 +/- 0.4 pmol of catechol/min/nmol of P450) may be involved in etoposide metabolism at therapeutic concentrations of free drug. Etoposide 94-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-73 15365903-1 2004 Density functional theoretical studies of monooxygenation reactivity of the high-valent oxoiron(IV) porphyrin cation-radical compound of cytochrome P450, the so-called Compound I, and of its precursor, the ferric(III)-hydroperoxide species, are described. porphyrin cation-radical 100-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 15365903-5 2004 If and when Compound I is absent, P450 oxidation will logically proceed by another form, but this has to be more reactive than ferric(III)-hydroperoxide. ferric(iii)-hydroperoxide 127-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 15121764-0 2004 Metabolism of N,N",N"-triethylenethiophosphoramide by CYP2B1 and CYP2B6 results in the inactivation of both isoforms by two distinct mechanisms. Thiotepa 14-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 15121764-1 2004 The anticancer drug N,N,"N"-triethylenethiophosphoramide (tTEPA) inactivated CYP2B6 and CYP2B1 in the reconstituted system in a time-, concentration-, and NADPH-dependent manner indicative of mechanism-based inactivation. Thiotepa 20-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 15121764-1 2004 The anticancer drug N,N,"N"-triethylenethiophosphoramide (tTEPA) inactivated CYP2B6 and CYP2B1 in the reconstituted system in a time-, concentration-, and NADPH-dependent manner indicative of mechanism-based inactivation. Thiotepa 58-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 15121764-8 2004 However, unlike CYP2B1, the tTEPA-inactivated human isoform showed losses in the cytochrome P450 (P450) CO spectrum, the pyridine hemochrome spectrum, and in the amount of native heme that were comparable with the loss in the 7-EFC and benzphetamine activity, suggesting that activity loss was brought about by a tTEPA-reactive intermediate damaging the CYP2B6 heme. Thiotepa 28-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 354-360 15284537-0 2004 Pharmacogenetic roles of CYP2C19 and CYP2B6 in the metabolism of R- and S-mephobarbital in humans. r- and s-mephobarbital 65-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 15279838-1 2004 We newly developed 10 Salmonela typhimurium TA1538 strains each co-expressing a form of human cytochrome P450s (P450 or CYP) together with NADPH-cytochrome P450 reductase (CPR) for highly sensitive detection of mutagenic activation of mycotoxins, polycyclic aromatic hydrocarbons, heterocyclic amines, and aromatic amines at low substrate concentrations. Polycyclic Aromatic Hydrocarbons 247-279 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-123 15279838-1 2004 We newly developed 10 Salmonela typhimurium TA1538 strains each co-expressing a form of human cytochrome P450s (P450 or CYP) together with NADPH-cytochrome P450 reductase (CPR) for highly sensitive detection of mutagenic activation of mycotoxins, polycyclic aromatic hydrocarbons, heterocyclic amines, and aromatic amines at low substrate concentrations. heterocyclic amines 281-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-123 15279838-1 2004 We newly developed 10 Salmonela typhimurium TA1538 strains each co-expressing a form of human cytochrome P450s (P450 or CYP) together with NADPH-cytochrome P450 reductase (CPR) for highly sensitive detection of mutagenic activation of mycotoxins, polycyclic aromatic hydrocarbons, heterocyclic amines, and aromatic amines at low substrate concentrations. aromatic amines 306-321 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-123 15294351-1 2004 The effects of polychlorinated biphenyls (PCBs) on human cytochrome P450 aromatase activity in vitro were investigated using a commercially available microsomal fraction obtained from baculovirus infected insects that had been transfected with the human CYP19 gene and cytochrome P450 reductase. Polychlorinated Biphenyls 42-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 15284537-1 2004 OBJECTIVES AND METHODS: We assessed the relationship between the metabolism of R- and S-mephobarbital (MPB) and genetic polymorphisms of cytochrome P450 (CYP) 2C19 and CYP2B6. r- and s-mephobarbital 79-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 168-174 15284537-1 2004 OBJECTIVES AND METHODS: We assessed the relationship between the metabolism of R- and S-mephobarbital (MPB) and genetic polymorphisms of cytochrome P450 (CYP) 2C19 and CYP2B6. Mephobarbital 103-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 168-174 15194512-1 2004 Efavirenz (EFV) is metabolized by cytochrome P450 2B6 (CYP2B6) in the liver. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-53 15337633-7 2004 Inhibition of P450 and other cytochromes by fluconazole may incapacitate Borrelia upon longterm exposure. Fluconazole 44-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 15194512-1 2004 Efavirenz (EFV) is metabolized by cytochrome P450 2B6 (CYP2B6) in the liver. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 15194512-1 2004 Efavirenz (EFV) is metabolized by cytochrome P450 2B6 (CYP2B6) in the liver. efavirenz 11-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-53 15194512-1 2004 Efavirenz (EFV) is metabolized by cytochrome P450 2B6 (CYP2B6) in the liver. efavirenz 11-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 15180495-4 2004 With in vivo animal models, acute moderate hypoxia (PaO2 of around 35-50 mm Hg) reduces the clearance of drugs biotransformed by CYP1A1, CYP1A2, CYP2B6, CYP2C9, CYP2C19 and CYP2E1, although hypoxia does not affect the clearance of drugs biotransformed by CYP3A6. pao2 52-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-151 15123723-2 2004 Phenytoin (PHY), a widely used antiepileptic drug, is a potent inducer of CYP2B6 in primary human hepatocytes, but does not activate human pregnane X receptor (PXR) significantly in cell-based transfection assays at the same concentrations associated with potent induction of CYP2B6. Phenytoin 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 15123723-2 2004 Phenytoin (PHY), a widely used antiepileptic drug, is a potent inducer of CYP2B6 in primary human hepatocytes, but does not activate human pregnane X receptor (PXR) significantly in cell-based transfection assays at the same concentrations associated with potent induction of CYP2B6. Phenytoin 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 276-282 15123723-2 2004 Phenytoin (PHY), a widely used antiepileptic drug, is a potent inducer of CYP2B6 in primary human hepatocytes, but does not activate human pregnane X receptor (PXR) significantly in cell-based transfection assays at the same concentrations associated with potent induction of CYP2B6. Phenytoin 11-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 15123723-2 2004 Phenytoin (PHY), a widely used antiepileptic drug, is a potent inducer of CYP2B6 in primary human hepatocytes, but does not activate human pregnane X receptor (PXR) significantly in cell-based transfection assays at the same concentrations associated with potent induction of CYP2B6. Phenytoin 11-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 276-282 15123723-4 2004 In primary human hepatocytes, expression of CYP2B6 reporter genes containing phenobarbital-responsive enhancer module (PBREM) or PBREM/xenobiotic-responsive enhancer module (XREM) response elements were activated up to 14- and 28-fold, respectively, by 50 microm PHY. Phenobarbital 77-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 15158752-0 2004 Omega-hydroxylation of farnesol by mammalian cytochromes p450. Farnesol 23-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 15158752-1 2004 Studies have shown that mammalian cytochromes p450 participate in the metabolism of terpenes, yet their role in the biotransformation of farnesol, an endogenous 15-carbon isoprenol, is unknown. Terpenes 84-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-50 15158752-1 2004 Studies have shown that mammalian cytochromes p450 participate in the metabolism of terpenes, yet their role in the biotransformation of farnesol, an endogenous 15-carbon isoprenol, is unknown. Farnesol 137-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-50 15158752-6 2004 Among a series of available human p450 enzymes, only CYP2C19 also produced 12-hydroxyfarnesol. 12-hydroxyfarnesol 75-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 15248218-9 2004 In a survival analysis, patients homozygous for CYP2B6*5 (n = 3) or CYP2C19*2 (n = 4) had a higher probability of reaching ESRD (P = 0.0005) and of doubling the creatinine level (P = 0.0005) as well as a trend toward a lower probability of achieving a complete renal response (P = 0.051). Creatinine 161-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-54 15155554-0 2004 Selective inhibition of CYP2B6-catalyzed bupropion hydroxylation in human liver microsomes in vitro. Bupropion 41-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 15167626-1 2004 Efavirenz is a drug subject to extensive metabolism, mainly by the cytochrome P-450 isoenzyme CYP2B6, known to exhibit extensive interindividual variability. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 15155554-2 2004 The goal of this work was to find a selective and potent chemical in vitro inhibitor toward CYP2B6 using bupropion hydroxylation as a model reaction. Bupropion 105-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 15155554-4 2004 Metyrapone, xanthate C8, and benzylisothiocyanate inhibited several other cytochrome P450 activities rather effectively, some of them even more potently than CYP2B6, and consequently are unsuitable as CYP2B6-selective probes. Metyrapone 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-164 15155554-4 2004 Metyrapone, xanthate C8, and benzylisothiocyanate inhibited several other cytochrome P450 activities rather effectively, some of them even more potently than CYP2B6, and consequently are unsuitable as CYP2B6-selective probes. Metyrapone 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 201-207 15155554-4 2004 Metyrapone, xanthate C8, and benzylisothiocyanate inhibited several other cytochrome P450 activities rather effectively, some of them even more potently than CYP2B6, and consequently are unsuitable as CYP2B6-selective probes. ethylxanthate 12-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-164 15155554-4 2004 Metyrapone, xanthate C8, and benzylisothiocyanate inhibited several other cytochrome P450 activities rather effectively, some of them even more potently than CYP2B6, and consequently are unsuitable as CYP2B6-selective probes. benzyl isothiocyanate 29-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-164 15155554-6 2004 The inhibition type of ticlopidine was found to be mixed type, with a component of mechanism-based inhibition, whereas thioTEPA inhibited CYP2B6 in a competitive manner. Thiotepa 119-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 15155554-8 2004 For thioTEPA the next sensitive P450 activity after CYP2B6 was coumarin 7-hydroxylation (IC(50), 256 microM). Thiotepa 4-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 15155554-8 2004 For thioTEPA the next sensitive P450 activity after CYP2B6 was coumarin 7-hydroxylation (IC(50), 256 microM). coumarin 63-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 15155554-10 2004 Thus, thioTEPA is a drug of choice when high CYP2B6 selectivity among major P450 enzymes is required. Thiotepa 6-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 15277012-0 2004 Quantitative structure-activity relationships within a homologous series of 7-alkoxyresorufins exhibiting activity towards CYP1A and CYP2B enzymes: molecular modelling studies on key members of the resorufin series with CYP2C5-derived models of human CYP1A1, CYP1A2, CYP2B6 and CYP3A4. 7-alkoxyresorufins 76-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-138 15277012-0 2004 Quantitative structure-activity relationships within a homologous series of 7-alkoxyresorufins exhibiting activity towards CYP1A and CYP2B enzymes: molecular modelling studies on key members of the resorufin series with CYP2C5-derived models of human CYP1A1, CYP1A2, CYP2B6 and CYP3A4. 7-alkoxyresorufins 76-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 267-273 15277012-7 2004 A quadratic relationship between compound lipophilicity and binding to CYP2B enzymes was apparent, and which indicated maximal interaction for 7-pentoxyresorufin. pentoxyresorufin 143-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-76 15081898-0 2004 The mechanism-based inactivation of p450 2B4 by tert-butyl 1-methyl-2-propynyl ether: structural determination of the adducts to the p450 heme. tert-butyl 1-methyl-2-propynyl ether 48-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 15277012-10 2004 Calculation of the binding affinities for methoxy-, ethoxy-, pentoxy- and benzyloxy-resorufins towards either CYP1A2 or CYP2B6 enzymes, depending on the 7-alkoxyresorufin agree favourably with experimental values obtained from K(m) determinations. methoxy-, ethoxy-, pentoxy- and benzyloxy-resorufins 42-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 15277012-10 2004 Calculation of the binding affinities for methoxy-, ethoxy-, pentoxy- and benzyloxy-resorufins towards either CYP1A2 or CYP2B6 enzymes, depending on the 7-alkoxyresorufin agree favourably with experimental values obtained from K(m) determinations. 7-alkoxyresorufin 153-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 15081432-0 2004 Inhibition of human P450 enzymes by nicotinic acid and nicotinamide. Niacin 36-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 15081432-0 2004 Inhibition of human P450 enzymes by nicotinic acid and nicotinamide. Niacinamide 55-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 15081432-3 2004 Based on their common pyridine functionality, we hypothesized that these two molecules could inhibit human P450 enzymes. pyridine 22-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-111 15081898-0 2004 The mechanism-based inactivation of p450 2B4 by tert-butyl 1-methyl-2-propynyl ether: structural determination of the adducts to the p450 heme. tert-butyl 1-methyl-2-propynyl ether 48-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 15081898-0 2004 The mechanism-based inactivation of p450 2B4 by tert-butyl 1-methyl-2-propynyl ether: structural determination of the adducts to the p450 heme. Heme 138-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 15081898-0 2004 The mechanism-based inactivation of p450 2B4 by tert-butyl 1-methyl-2-propynyl ether: structural determination of the adducts to the p450 heme. Heme 138-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 15081898-1 2004 tert-Butyl 1-methyl-2-propynyl ether (tBMP) was analyzed for its ability to act as a mechanism-based inactivator of p450 2B4. tert-butyl 1-methyl-2-propynyl ether 0-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-120 15081898-1 2004 tert-Butyl 1-methyl-2-propynyl ether (tBMP) was analyzed for its ability to act as a mechanism-based inactivator of p450 2B4. tert-butyl 1-methyl-2-propynyl ether 38-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-120 15081898-2 2004 tBMP inactivated p450 2B4 in a time-, concentration-, and NADPH-dependent manner. tert-butyl 1-methyl-2-propynyl ether 0-4 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 15081898-2 2004 tBMP inactivated p450 2B4 in a time-, concentration-, and NADPH-dependent manner. NADP 58-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 15133245-7 2004 We also examined whether oxatomide showed inhibitory effects on metabolic activity of individual P450 isozymes using model substrates for each isozyme. oxatomide 25-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-101 15133245-0 2004 Identification of human p450 isoforms involved in the metabolism of the antiallergic drug, oxatomide, and its inhibitory effect on enzyme activity. oxatomide 91-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 15081898-3 2004 Losses in activity occurred with concurrent losses in the reduced CO spectrum and native p450 heme; however, there was a greater loss in activity than could be accounted for by reduced CO spectra or native heme loss. Heme 94-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-93 15133245-4 2004 On the other hand, isoforms of P450 involved in the metabolism of oxatomide have not been clarified. oxatomide 66-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 15081898-7 2004 Proton NMR studies suggest that the two modified hemes in p450 2B1 are N-alkylated on pyrrole rings A and D. Heme 49-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 15081898-7 2004 Proton NMR studies suggest that the two modified hemes in p450 2B1 are N-alkylated on pyrrole rings A and D. Pyrroles 86-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 15170127-2 2004 Characterization of the E71A mutant protein of p450nor highlights the existence of a unique mechanism for binding NADH that depends on the salt bridge network between Glu71, Arg64 and Asp88. NAD 114-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 15144223-0 2004 Enzymes involved in the metabolism of the carcinogen 2-nitroanisole: evidence for its oxidative detoxication by human cytochromes P450. 2-nitroanisole 53-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-134 15144223-11 2004 The role of specific P450 enzymes in the metabolism of 2-NA in human hepatic microsomes was investigated by correlating specific P450-dependent reactions with the levels of 2-NA metabolites formed by the same microsomes and by examining the effects of specific inhibitors of P450 enzymes on 2-NA metabolism. 2-nitroanisole 55-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 15144223-11 2004 The role of specific P450 enzymes in the metabolism of 2-NA in human hepatic microsomes was investigated by correlating specific P450-dependent reactions with the levels of 2-NA metabolites formed by the same microsomes and by examining the effects of specific inhibitors of P450 enzymes on 2-NA metabolism. 2-nitroanisole 55-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 15144223-11 2004 The role of specific P450 enzymes in the metabolism of 2-NA in human hepatic microsomes was investigated by correlating specific P450-dependent reactions with the levels of 2-NA metabolites formed by the same microsomes and by examining the effects of specific inhibitors of P450 enzymes on 2-NA metabolism. 2-nitroanisole 55-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 15144223-13 2004 These results, the first report on the metabolism of 2-NA by human P450 enzymes, clearly demonstrate that P450 2E1 is the major human enzyme oxidizing this carcinogen in human liver. 2-nitroanisole 53-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-71 15144223-13 2004 These results, the first report on the metabolism of 2-NA by human P450 enzymes, clearly demonstrate that P450 2E1 is the major human enzyme oxidizing this carcinogen in human liver. 2-nitroanisole 53-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-110 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 2 subfamily B member 6 Homo sapiens 15-19 15370961-3 2004 S- and R-etodolac were converted to the acylglucuronide and hydroxylated metabolites by UDP-glucuronosyltransferase (UGT) and cytochrome P450 in microsomes. s- and r-etodolac 0-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-141 15370961-3 2004 S- and R-etodolac were converted to the acylglucuronide and hydroxylated metabolites by UDP-glucuronosyltransferase (UGT) and cytochrome P450 in microsomes. acylglucuronide 40-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-141 15370961-6 2004 In contrast, R-etodolac was hydroxylated preferentially than S-etodolac by P450. Etodolac 13-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 15370961-6 2004 In contrast, R-etodolac was hydroxylated preferentially than S-etodolac by P450. Etodolac 61-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 15370961-8 2004 Of several human P450 enzymes, CYP2C9 had the greatest activity for hydroxylation of R-etodolac. Etodolac 85-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 15078194-5 2004 Metabolic activation occurs by N-hydroxylation, a reaction catalyzed by cytochromes p450 (CYP). Nitrogen 31-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 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 2 subfamily B member 6 Homo sapiens 37-41 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. NADP 31-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-41 15089095-2 2004 The oxidation rate of NADPH by p450 1A2/NPR increased with time in the presence of IQ until depletion of NADPH. NADP 22-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 15089095-2 2004 The oxidation rate of NADPH by p450 1A2/NPR increased with time in the presence of IQ until depletion of NADPH. NADP 105-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 15228172-5 2004 The potential of applying pharmacogenetic screening before therapy in the treatment of cancer seems to be greatest for CYP2B6 (cyclophosphamide treatment), CYP2C8 (paclitaxel therapy), and CYP3A5; however, the drugs of interest still need to be identified for this latter enzyme. Cyclophosphamide 127-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 15083067-0 2004 Pharmacogenetic determinants of interindividual variability in bupropion hydroxylation by cytochrome P450 2B6 in human liver microsomes. Bupropion 63-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-109 15083067-1 2004 Bupropion is primarily metabolized in human liver by cytochrome P450 (CYP) 2B6, an isoform that shows high interindividual variability in expression and catalysis. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-78 15083067-8 2004 A consideration of the effects of these variables on CYP2B6 mRNA and protein levels suggests that alcohol use is associated with enhanced CYP2B6 gene transcription, but the presence of at least one *6B allele reduces this effect on bupropion hydroxylation at the post-transcriptional level. Alcohols 98-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 15083067-8 2004 A consideration of the effects of these variables on CYP2B6 mRNA and protein levels suggests that alcohol use is associated with enhanced CYP2B6 gene transcription, but the presence of at least one *6B allele reduces this effect on bupropion hydroxylation at the post-transcriptional level. Alcohols 98-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 15083067-9 2004 In conclusion, the results of this study indicate that interindividual variability in bupropion hydroxylation is a consequence of interactions between environmental and genetic influences on CYP2B6 gene function. Bupropion 86-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 191-197 15123254-2 2004 In this issue of Chemistry & Biology, Arnold and coworkers report their use of the SCHEMA algorithm to effectively predict ideal hybrids of cytochromes p450. Adenosine Monophosphate 28-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 156-160 14973583-1 2004 Novel cyclopropyl containing fatty acids are good substrates for P450(BM3) catalysed hydroxylation and analysis of their oxidation products indicates the presence of a radical intermediate (maximum rebound rate 2.6 x 10(10) s(-1)) and the absence of any cationic intermediate. cyclopropyl containing fatty acids 6-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-73 14977870-4 2004 CYP2B6 and CYP3A4 protein and activities were assessed by Western blotting, bupropion hydroxylation, and testosterone 6beta-hydroxylation, respectively. Bupropion 76-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 14977870-4 2004 CYP2B6 and CYP3A4 protein and activities were assessed by Western blotting, bupropion hydroxylation, and testosterone 6beta-hydroxylation, respectively. Testosterone 105-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Clotrimazole 47-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Phenobarbital 61-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Phenytoin 76-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Rifampin 91-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Clotrimazole 182-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Phenytoin 196-205 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 14977870-7 2004 EC50 values for CYP2B6 and CYP3A4 induction by clotrimazole, phenobarbital, phenytoin, and rifampin were strongly correlated (r2 = 0.99) and were statistically indistinguishable for clotrimazole, phenytoin, and rifampin. Rifampin 211-219 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 14977870-8 2004 Kinetic constants governing time-dependent induction by phenobarbital and rifampin were also similar between CYP2B6 and CYP3A4. Rifampin 74-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 14757833-10 2004 Applying the guidelines to find oligonucleotide microarray probes for P450 genes, we confirmed the ability of our point mutation method to differentiate the individual genes in terms of thermodynamic calculations of hybridization and sequence similarity. Oligonucleotides 32-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 14978227-1 2004 Human constitutive androstane (or active) receptor (hCAR), a member of the nuclear receptor superfamily NR1I3, regulates the expression of several genes that are mainly involved in the metabolism of endogenous and xenobiotic compounds (e.g., CYP2B6, CYP3A4, and UGT1A1). androstane 19-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-248 14978227-7 2004 In the reporter gene assays employing the phenobarbital-responsible enhancer module (PBREM) from CYP2B6 and UGT1A1 genes, the splice variants, except for SV1, were inactive, whereas SV1 transactivated the CYP2B6 PBREM but not the UGT1A1 PBREM reporter. Phenobarbital 42-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 14978227-7 2004 In the reporter gene assays employing the phenobarbital-responsible enhancer module (PBREM) from CYP2B6 and UGT1A1 genes, the splice variants, except for SV1, were inactive, whereas SV1 transactivated the CYP2B6 PBREM but not the UGT1A1 PBREM reporter. Phenobarbital 42-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 205-211 14977851-0 2004 The metabolism of pyrazoloacridine (NSC 366140) by cytochromes p450 and flavin monooxygenase in human liver microsomes. NSC 366140 18-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-92 14718257-4 2004 Recombinant human CYP2B6, 2C9 and 2C19 biotransformed 5-HT in 5-hydroxyindol acetic acid, but not CYP1A2, 2D6 or 3A4. Hydroxytryptophol 62-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 14718257-6 2004 In the presence of CYP2B6, 2C9 and 2C19, 5-HT relaxed precontracted isolated aortic rings, with or without endothelium, an effect prevented by the addition of methylene blue and an inhibitor of catalase, but not by myoglobin. Methylene Blue 159-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 14718257-8 2004 In conclusion, CYP2B6, 2C9 and 2C19 biotransform 5-HT, yielding hydroxylamine, which is converted to nitric oxide in the presence of catalase. Hydroxylamine 64-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 14718257-8 2004 In conclusion, CYP2B6, 2C9 and 2C19 biotransform 5-HT, yielding hydroxylamine, which is converted to nitric oxide in the presence of catalase. Nitric Oxide 101-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 15034803-3 2004 In response to stimuli, arachidonic acid is mobilized from phospholipid pools and metabolized by cyclooxygenases (COX), lipoxygenases (LOX), and p450 epoxygenases (EOX) to form a variety of eicosanoids. Eicosanoids 190-201 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-149 15034803-3 2004 In response to stimuli, arachidonic acid is mobilized from phospholipid pools and metabolized by cyclooxygenases (COX), lipoxygenases (LOX), and p450 epoxygenases (EOX) to form a variety of eicosanoids. Arachidonic Acid 24-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-149 14709076-0 2004 Hydroxylation by the hydroperoxy-iron species in cytochrome P450 enzymes. hydroperoxy-iron 21-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-64 14709076-1 2004 Intramolecular and intermolecular kinetic isotope effects (KIEs) were determined for hydroxylation of the enantiomers of trans-2-(p-trifluoromethylphenyl)cyclopropylmethane (1) by hepatic cytochrome P450 enzymes, P450s 2B1, Delta2B4, Delta2B4 T302A, Delta2E1, and Delta2E1 T303A. trans-2-(p-trifluoromethylphenyl)cyclopropylmethane 121-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-203 14709076-8 2004 The intra- and intermolecular KIE results were combined to determine the relative rate constants for the unmasking and hydroxylation reactions, and a qualitative correlation was found for the unmasking reaction and release of hydrogen peroxide from four of the P450 enzymes in the absence of substrate. Hydrogen Peroxide 226-243 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 261-265 14729637-4 2004 Retrovirus encoding a CYP2B6-IRES-P450R expression cassette was shown to induce strong P450-dependent CPA cytotoxicity in a population of infected 9L gliosarcoma cells. Cyclophosphamide 102-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 15509185-12 2004 The O-demethylation of tramadol to M1, the main analgesic effective metabolite, is catalysed by cytochrome P450 (CYP) 2D6, whereas N-demethylation to M2 is catalysed by CYP2B6 and CYP3A4. Tramadol 23-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 14729637-5 2004 Adeno-P450, a replication-defective, E1/E3 region-deleted adenovirus engineered to express CYP2B6-IRES-P450R, induced intracellular CPA 4-hydroxylation, and CPA cytotoxicity, in a broad range of human cancer cell lines. Cyclophosphamide 132-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 15122651-10 2004 The recombinant expressed human CYP2F1 enzyme had a K(m) value of 3.83 microM and a V(max) value of 0.01 pmol/pmol p450 enzyme/min demonstrating a reasonably efficient catalysis of benzene metabolism (V(max)/K(m) = 2.6). Benzene 181-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-119 14709627-5 2004 The reference inhibitor ketoconazole was less selective and exhibited potent inhibition (IC(50) values <10 microM) for CYP1A1, CYP1B1, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP4F2, and CYP4F12. Ketoconazole 24-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 15508429-5 2004 For example, in most cases it would appear that there is a set number of intervening "heavy" atoms (atoms other than hydrogen) between sites of metabolism and key hydrogen bond acceptors (or donors) for human P450 substrates, with the number of intervening atoms being dependent upon the type of P450 involved. Hydrogen 117-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 209-213 15508429-5 2004 For example, in most cases it would appear that there is a set number of intervening "heavy" atoms (atoms other than hydrogen) between sites of metabolism and key hydrogen bond acceptors (or donors) for human P450 substrates, with the number of intervening atoms being dependent upon the type of P450 involved. Hydrogen 117-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 296-300 15508429-5 2004 For example, in most cases it would appear that there is a set number of intervening "heavy" atoms (atoms other than hydrogen) between sites of metabolism and key hydrogen bond acceptors (or donors) for human P450 substrates, with the number of intervening atoms being dependent upon the type of P450 involved. Hydrogen 163-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 209-213 15508429-5 2004 For example, in most cases it would appear that there is a set number of intervening "heavy" atoms (atoms other than hydrogen) between sites of metabolism and key hydrogen bond acceptors (or donors) for human P450 substrates, with the number of intervening atoms being dependent upon the type of P450 involved. Hydrogen 163-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 296-300 15122651-3 2004 The toxic effects of benzene are dependent on its metabolism by the cytochrome p450 enzyme system. Benzene 21-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 14628297-5 2004 Only CYP2B6, 2C19, and 3A4 generated measurable EDDP from 1 microg/ml of racemic methadone. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 48-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-11 14628297-5 2004 Only CYP2B6, 2C19, and 3A4 generated measurable EDDP from 1 microg/ml of racemic methadone. Methadone 81-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-11 14628297-6 2004 The hierarchy of EDDP generation was CYP2B6 > CYP2C19 >/= CYP3A4. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 17-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 14628297-7 2004 At 10 microg/ml of methadone, CYP2C9 and CYP2D6 also generated EDDP, but in at least 10-fold lower quantities than CYP2B6. Methadone 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 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 2 subfamily B member 6 Homo sapiens 48-54 15122651-4 2004 The cytochrome p450 enzymes CYP2E1 and CYP2F2 are the major contributors to the bioactivation of benzene in rats and mice. Benzene 97-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 15122651-11 2004 Thus, these studies have demonstrated in human lung cell lines that benzene is bioactivated by two lung-expressed p450 enzymes. Benzene 68-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-118 14683479-6 2003 Several recent findings, including discovery of a human CAR specific activator 6-(4-chlorophenyl:imidazo[2,1-b]thiazole-5carbaldehyde O-(3,4-dichlorobenzyl)oxime (CITCO), identification of a far-distal xenobiotic-responsive enhancer module (XREM) in the CYP2B6 gene promoter, and generation of CAR-null mice as a model of characterizing CAR target gene expression, will also be discussed. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 163-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 254-260 14563790-0 2004 Potent mechanism-based inhibition of human CYP2B6 by clopidogrel and ticlopidine. Clopidogrel 53-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 14563790-0 2004 Potent mechanism-based inhibition of human CYP2B6 by clopidogrel and ticlopidine. Ticlopidine 69-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 14563790-4 2004 Both clopidogrel and ticlopidine inhibited CYP2B6 with highest potency and CYP2C19 with lower potency. Clopidogrel 5-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 14563790-4 2004 Both clopidogrel and ticlopidine inhibited CYP2B6 with highest potency and CYP2C19 with lower potency. Ticlopidine 21-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 14563790-6 2004 Inhibition of CYP2B6 was time- and concentration-dependent, and as shown by dialysis experiments, it was irreversible and dependent on NADPH, suggesting a mechanism-based mode of action. NADP 135-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 14563790-9 2004 A chemical mechanism is discussed based on the known metabolic activation of clopidogrel and on the finding that hemoprotein integrity of recombinant CYP2B6 was not affected by irreversible inhibition. Clopidogrel 77-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 14563790-10 2004 These results suggest the possibility of drug interactions between thienopyridine derivates and drug substrates of CYP2B6 and CYP2C19. thienopyridine 67-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 14640711-5 2003 The system was used both to yield the Michaelis constant (K(m)) of the P450 biotransformation of imipramine into desipramine and to determine the IC50 value of a chemical inhibitor (tranylcypromine) for this CYP2C19-mediated reaction. Imipramine 97-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 14640711-5 2003 The system was used both to yield the Michaelis constant (K(m)) of the P450 biotransformation of imipramine into desipramine and to determine the IC50 value of a chemical inhibitor (tranylcypromine) for this CYP2C19-mediated reaction. Desipramine 113-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 14640711-5 2003 The system was used both to yield the Michaelis constant (K(m)) of the P450 biotransformation of imipramine into desipramine and to determine the IC50 value of a chemical inhibitor (tranylcypromine) for this CYP2C19-mediated reaction. Tranylcypromine 182-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 14628297-9 2004 In human liver microsomes with high and low CYP2B6 expression but equivalent CYP3A4 expression, high CYP2B6 expression microsomes generated twice the amount of EDDP from 10 microg/ml of methadone than low CYP2B6 expression microsomes. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 160-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 14628297-9 2004 In human liver microsomes with high and low CYP2B6 expression but equivalent CYP3A4 expression, high CYP2B6 expression microsomes generated twice the amount of EDDP from 10 microg/ml of methadone than low CYP2B6 expression microsomes. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 160-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 14628297-9 2004 In human liver microsomes with high and low CYP2B6 expression but equivalent CYP3A4 expression, high CYP2B6 expression microsomes generated twice the amount of EDDP from 10 microg/ml of methadone than low CYP2B6 expression microsomes. Methadone 186-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 14628297-9 2004 In human liver microsomes with high and low CYP2B6 expression but equivalent CYP3A4 expression, high CYP2B6 expression microsomes generated twice the amount of EDDP from 10 microg/ml of methadone than low CYP2B6 expression microsomes. Methadone 186-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 14628297-10 2004 When stereoselective metabolism of racemic methadone by CYP2B6, 2C19, and 3A4 was examined using an enantiospecific methadone assay, CYP2B6 preferentially metabolized (S)-methadone, CYP2C19 preferentially metabolized (R)-methadone, and CYP3A4 showed no preference. Methadone 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 14628297-10 2004 When stereoselective metabolism of racemic methadone by CYP2B6, 2C19, and 3A4 was examined using an enantiospecific methadone assay, CYP2B6 preferentially metabolized (S)-methadone, CYP2C19 preferentially metabolized (R)-methadone, and CYP3A4 showed no preference. Methadone 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 14628297-10 2004 When stereoselective metabolism of racemic methadone by CYP2B6, 2C19, and 3A4 was examined using an enantiospecific methadone assay, CYP2B6 preferentially metabolized (S)-methadone, CYP2C19 preferentially metabolized (R)-methadone, and CYP3A4 showed no preference. Methadone 116-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 14628297-10 2004 When stereoselective metabolism of racemic methadone by CYP2B6, 2C19, and 3A4 was examined using an enantiospecific methadone assay, CYP2B6 preferentially metabolized (S)-methadone, CYP2C19 preferentially metabolized (R)-methadone, and CYP3A4 showed no preference. Methadone 116-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 14628297-10 2004 When stereoselective metabolism of racemic methadone by CYP2B6, 2C19, and 3A4 was examined using an enantiospecific methadone assay, CYP2B6 preferentially metabolized (S)-methadone, CYP2C19 preferentially metabolized (R)-methadone, and CYP3A4 showed no preference. Methadone 167-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 14628297-10 2004 When stereoselective metabolism of racemic methadone by CYP2B6, 2C19, and 3A4 was examined using an enantiospecific methadone assay, CYP2B6 preferentially metabolized (S)-methadone, CYP2C19 preferentially metabolized (R)-methadone, and CYP3A4 showed no preference. Methadone 167-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 14628297-10 2004 When stereoselective metabolism of racemic methadone by CYP2B6, 2C19, and 3A4 was examined using an enantiospecific methadone assay, CYP2B6 preferentially metabolized (S)-methadone, CYP2C19 preferentially metabolized (R)-methadone, and CYP3A4 showed no preference. Methadone 217-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 14628297-10 2004 When stereoselective metabolism of racemic methadone by CYP2B6, 2C19, and 3A4 was examined using an enantiospecific methadone assay, CYP2B6 preferentially metabolized (S)-methadone, CYP2C19 preferentially metabolized (R)-methadone, and CYP3A4 showed no preference. Methadone 217-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 14628297-11 2004 These data suggest that multiple CYPs metabolized methadone but CYP2B6 had the highest V(max)/K(m). Methadone 50-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 14628297-13 2004 Our data could explain why the plasma concentration ratio of R/S methadone is variable and why drugs that induce CYP2B6 such as nevirapine and efavirenz also induce methadone metabolism, while the CYP3A4 inducer rifabutin has no effect on methadone pharmacokinetics. Methadone 65-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 14628297-13 2004 Our data could explain why the plasma concentration ratio of R/S methadone is variable and why drugs that induce CYP2B6 such as nevirapine and efavirenz also induce methadone metabolism, while the CYP3A4 inducer rifabutin has no effect on methadone pharmacokinetics. Nevirapine 128-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 14628297-13 2004 Our data could explain why the plasma concentration ratio of R/S methadone is variable and why drugs that induce CYP2B6 such as nevirapine and efavirenz also induce methadone metabolism, while the CYP3A4 inducer rifabutin has no effect on methadone pharmacokinetics. efavirenz 143-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 14628297-13 2004 Our data could explain why the plasma concentration ratio of R/S methadone is variable and why drugs that induce CYP2B6 such as nevirapine and efavirenz also induce methadone metabolism, while the CYP3A4 inducer rifabutin has no effect on methadone pharmacokinetics. Methadone 165-174 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 14628297-13 2004 Our data could explain why the plasma concentration ratio of R/S methadone is variable and why drugs that induce CYP2B6 such as nevirapine and efavirenz also induce methadone metabolism, while the CYP3A4 inducer rifabutin has no effect on methadone pharmacokinetics. Rifabutin 212-221 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 14628297-13 2004 Our data could explain why the plasma concentration ratio of R/S methadone is variable and why drugs that induce CYP2B6 such as nevirapine and efavirenz also induce methadone metabolism, while the CYP3A4 inducer rifabutin has no effect on methadone pharmacokinetics. Methadone 165-174 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-119 14727741-0 2004 Biotransformation of atrazine in transgenic tobacco cell culture expressing human P450. Atrazine 21-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 14683479-1 2003 Cytochrome p450 2B genes have been used extensively as prototypical models to study phenobarbital induction of p450 enzymes. Phenobarbital 84-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 14683479-1 2003 Cytochrome p450 2B genes have been used extensively as prototypical models to study phenobarbital induction of p450 enzymes. Phenobarbital 84-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-115 14683479-6 2003 Several recent findings, including discovery of a human CAR specific activator 6-(4-chlorophenyl:imidazo[2,1-b]thiazole-5carbaldehyde O-(3,4-dichlorobenzyl)oxime (CITCO), identification of a far-distal xenobiotic-responsive enhancer module (XREM) in the CYP2B6 gene promoter, and generation of CAR-null mice as a model of characterizing CAR target gene expression, will also be discussed. 6-(4-chlorophenyl: 79-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 254-260 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 2 subfamily B member 6 Homo sapiens 114-120 14563924-3 2003 A 1.6-A crystal structure of P450 2B4 reveals a large open cleft that extends from the protein surface directly to the heme iron between the alpha-helical and beta-sheet domains without perturbing the overall P450 fold. Heme 119-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 14563924-3 2003 A 1.6-A crystal structure of P450 2B4 reveals a large open cleft that extends from the protein surface directly to the heme iron between the alpha-helical and beta-sheet domains without perturbing the overall P450 fold. Iron 124-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 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 2 subfamily B member 6 Homo sapiens 212-218 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 2 subfamily B member 6 Homo sapiens 212-218 14678248-12 2003 Genotyping of patients with regards to CYP2B6, CYP2C9 and CYP2D6 may play a role in prediction of Z-4-OH-tam formation and, consequently, ultimate therapeutic benefit of tamoxifen treatment. z-4-oh-tam 98-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 14678248-12 2003 Genotyping of patients with regards to CYP2B6, CYP2C9 and CYP2D6 may play a role in prediction of Z-4-OH-tam formation and, consequently, ultimate therapeutic benefit of tamoxifen treatment. Tamoxifen 170-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 14632469-11 2003 The inferred active sites mapped out by the 4D-QSAR models suggest that hydrogen bond interactions are not prevalent when this class of P450 analogue inhibitors binds to the receptor active site. Hydrogen 72-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 14534519-0 2003 Inhibition of cytochrome P450 2B6 activity by hormone replacement therapy and oral contraceptive as measured by bupropion hydroxylation. Bupropion 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-33 14534519-1 2003 AIM: Our objective was to study the effect of hormone replacement therapy and a combination oral contraceptive on the cytochrome P450 (CYP) 2B6 activity with bupropion (INN, amfebutamone) hydroxylation used as a probe reaction. Bupropion 158-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-143 14534519-10 2003 CONCLUSIONS: Hormone replacement therapy markedly inhibited the CYP2B6-catalyzed hydroxylation of bupropion, whereas a combination oral contraceptive had only a modest effect on CYP2B6 activity. Bupropion 98-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 12975331-0 2003 Stereoselective metabolism of lansoprazole by human liver cytochrome P450 enzymes. Lansoprazole 30-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-73 12975331-1 2003 The stereoselective metabolism of lansoprazole enantiomers was evaluated by incubation of human liver microsomes and cDNA-expressed cytochrome p450 (p450) enzymes to understand and predict their stereoselective disposition in humans in vivo. Lansoprazole 34-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-147 12975331-1 2003 The stereoselective metabolism of lansoprazole enantiomers was evaluated by incubation of human liver microsomes and cDNA-expressed cytochrome p450 (p450) enzymes to understand and predict their stereoselective disposition in humans in vivo. Lansoprazole 34-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-153 12975331-5 2003 Total Clint values of hydroxy and sulfone metabolite formation catalyzed by all these p450 enzymes were consistently higher for S-lansoprazole than for the R-form. hydroxy and sulfone 22-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-90 12975331-5 2003 Total Clint values of hydroxy and sulfone metabolite formation catalyzed by all these p450 enzymes were consistently higher for S-lansoprazole than for the R-form. Lansoprazole 128-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-90 12975333-0 2003 In vitro metabolism studies on the isoxazole ring scission in the anti-inflammatory agent lefluonomide to its active alpha-cyanoenol metabolite A771726: mechanistic similarities with the cytochrome P450-catalyzed dehydration of aldoximes. Isoxazoles 35-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-202 12975333-0 2003 In vitro metabolism studies on the isoxazole ring scission in the anti-inflammatory agent lefluonomide to its active alpha-cyanoenol metabolite A771726: mechanistic similarities with the cytochrome P450-catalyzed dehydration of aldoximes. lefluonomide 90-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-202 12975333-11 2003 A mechanism for the p450-mediated ring scission is proposed in which the isoxazole ring nitrogen or oxygen coordinates to the reduced form of the heme followed by charge transfer from p450Fe(II) to the C=N bond or deprotonation of the C3-H, which results in a cleavage of the N-O bond. Isoxazoles 73-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 12975333-11 2003 A mechanism for the p450-mediated ring scission is proposed in which the isoxazole ring nitrogen or oxygen coordinates to the reduced form of the heme followed by charge transfer from p450Fe(II) to the C=N bond or deprotonation of the C3-H, which results in a cleavage of the N-O bond. Nitrogen 88-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 12975333-11 2003 A mechanism for the p450-mediated ring scission is proposed in which the isoxazole ring nitrogen or oxygen coordinates to the reduced form of the heme followed by charge transfer from p450Fe(II) to the C=N bond or deprotonation of the C3-H, which results in a cleavage of the N-O bond. Oxygen 100-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 12975333-11 2003 A mechanism for the p450-mediated ring scission is proposed in which the isoxazole ring nitrogen or oxygen coordinates to the reduced form of the heme followed by charge transfer from p450Fe(II) to the C=N bond or deprotonation of the C3-H, which results in a cleavage of the N-O bond. Heme 146-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 14515060-3 2003 Bupropion may be a probe drug for CYP2B6 activity in humans. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 14515060-0 2003 Bupropion and 4-OH-bupropion pharmacokinetics in relation to genetic polymorphisms in CYP2B6. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 14519119-3 2003 Here we report the redox properties of the cytochrome p450BM3 wild-type holoenzyme, and its isolated FAD reductase and p450 heme domains, when immobilized in a didodecyldimethylammonium bromide film cast on an edge-plane graphite electrode. didodecyldimethylammonium 160-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 14519119-3 2003 Here we report the redox properties of the cytochrome p450BM3 wild-type holoenzyme, and its isolated FAD reductase and p450 heme domains, when immobilized in a didodecyldimethylammonium bromide film cast on an edge-plane graphite electrode. Graphite 221-229 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 14515060-8 2003 Population kinetic analysis revealed that bupropion total clearance via CYP2B6 alleles *1, *2, *5 and *6 did not differ, but clearance via allele *4 was 1.66-fold higher compared to wild-type allele *1 (P=0.001). Bupropion 42-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 14515060-0 2003 Bupropion and 4-OH-bupropion pharmacokinetics in relation to genetic polymorphisms in CYP2B6. 4-oh-bupropion 14-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 14515060-9 2003 Corresponding to the high clearance of bupropion, carriers of the CYP2B6 genotype *1/*4 had significantly higher Cmax of hydroxybupropion compared to all other genotypes (P=0.03). Bupropion 39-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 14515060-2 2003 Genetic polymorphisms in cytochrome P450 2B6 (CYP2B6) may cause variability in bupropion pharmacokinetics since hydroxylation is known to be mediated by CYP2B6. Bupropion 79-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-44 14515060-2 2003 Genetic polymorphisms in cytochrome P450 2B6 (CYP2B6) may cause variability in bupropion pharmacokinetics since hydroxylation is known to be mediated by CYP2B6. Bupropion 79-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 14515060-9 2003 Corresponding to the high clearance of bupropion, carriers of the CYP2B6 genotype *1/*4 had significantly higher Cmax of hydroxybupropion compared to all other genotypes (P=0.03). hydroxybupropion 121-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 14515060-2 2003 Genetic polymorphisms in cytochrome P450 2B6 (CYP2B6) may cause variability in bupropion pharmacokinetics since hydroxylation is known to be mediated by CYP2B6. Bupropion 79-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 14515060-10 2003 Only a minor fraction of the variability in bupropion and hydroxybupropion kinetics could be explained by the known CYP2B6 amino acid variants, in particular by the CYP2B6*4 allele. Bupropion 44-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 14515060-10 2003 Only a minor fraction of the variability in bupropion and hydroxybupropion kinetics could be explained by the known CYP2B6 amino acid variants, in particular by the CYP2B6*4 allele. Bupropion 44-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-171 14515060-10 2003 Only a minor fraction of the variability in bupropion and hydroxybupropion kinetics could be explained by the known CYP2B6 amino acid variants, in particular by the CYP2B6*4 allele. hydroxybupropion 58-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 14515060-10 2003 Only a minor fraction of the variability in bupropion and hydroxybupropion kinetics could be explained by the known CYP2B6 amino acid variants, in particular by the CYP2B6*4 allele. hydroxybupropion 58-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-171 14515060-11 2003 The role of this allele should also be studied in other CYP2B6 substrates, including cyclophosphamide, halothane, mianserin, promethazine and propofol. Cyclophosphamide 85-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 12814665-1 2003 CYP2B6 metabolizes drugs such as nicotine and bupropion, and many toxins and carcinogens. Nicotine 33-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 12920164-1 2003 Selegiline was used as a model compound in a project aimed at comparing, evaluating, and integrating different in vitro approaches for the prediction of cytochrome p450 (p450)-catalyzed hepatic drug metabolism in humans (EUROCYP). Selegiline 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-168 12920164-1 2003 Selegiline was used as a model compound in a project aimed at comparing, evaluating, and integrating different in vitro approaches for the prediction of cytochrome p450 (p450)-catalyzed hepatic drug metabolism in humans (EUROCYP). Selegiline 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-174 12920164-8 2003 Homology modeling suggested the participation of CYP2B6 in the demethylation of selegiline and of CYP2D6 in the depropargylation of the drug. Selegiline 80-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 12920164-11 2003 In vitro, CYP2B6 was the most active form of p450 involved in selegiline metabolism. Selegiline 62-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 12920164-11 2003 In vitro, CYP2B6 was the most active form of p450 involved in selegiline metabolism. Selegiline 62-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-49 14604466-4 2003 CYP2B6 activity, protein and mRNA levels were increased by 2.5, 1.5 and 2.1 fold, respectively, by 20 microM rifampicin. Rifampin 109-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 14604466-5 2003 However, 10 microM bupropion minimally altered CYP2B6 (1.4, 1.1, 0.8 fold). Bupropion 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 12867484-0 2003 Identification of cytochrome p450 enzymes involved in the metabolism of 4"-methyl-alpha-pyrrolidinopropiophenone, a novel scheduled designer drug, in human liver microsomes. 4'-methyl-alpha-pyrrolidinopropiophenone 72-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 12867484-4 2003 The aim of the study presented here was to identify the human hepatic cytochrome p450 (p450) enzymes involved in the biotransformation of MPPP to 4"-hydroxymethyl-pyrrolidinopropiophenone. 4'-methyl-alpha-pyrrolidinopropiophenone 138-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 12867484-4 2003 The aim of the study presented here was to identify the human hepatic cytochrome p450 (p450) enzymes involved in the biotransformation of MPPP to 4"-hydroxymethyl-pyrrolidinopropiophenone. 4'-methyl-alpha-pyrrolidinopropiophenone 138-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 12867484-4 2003 The aim of the study presented here was to identify the human hepatic cytochrome p450 (p450) enzymes involved in the biotransformation of MPPP to 4"-hydroxymethyl-pyrrolidinopropiophenone. 4"-hydroxymethyl-pyrrolidinopropiophenone 146-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 12867484-4 2003 The aim of the study presented here was to identify the human hepatic cytochrome p450 (p450) enzymes involved in the biotransformation of MPPP to 4"-hydroxymethyl-pyrrolidinopropiophenone. 4"-hydroxymethyl-pyrrolidinopropiophenone 146-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 12867494-2 2003 In this study, (-)-verbenone was found to be converted to 10-hydroxyverbenone by rat and human liver microsomal cytochrome p450 (p450) enzymes. verbenone 15-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-127 12867494-2 2003 In this study, (-)-verbenone was found to be converted to 10-hydroxyverbenone by rat and human liver microsomal cytochrome p450 (p450) enzymes. verbenone 15-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 12867494-2 2003 In this study, (-)-verbenone was found to be converted to 10-hydroxyverbenone by rat and human liver microsomal cytochrome p450 (p450) enzymes. 10-Hydroxyverbenone 58-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-127 12867494-2 2003 In this study, (-)-verbenone was found to be converted to 10-hydroxyverbenone by rat and human liver microsomal cytochrome p450 (p450) enzymes. 10-Hydroxyverbenone 58-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 12867494-5 2003 Human recombinant CYP2A6 and CYP2B6 catalyzed (-)verbenone 10-hydroxylation at Vmax values of 15 and 21 nmol/min/nmol p450 with apparent Km values of 16 and 91 microM, respectively. verbenone 46-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 12867494-5 2003 Human recombinant CYP2A6 and CYP2B6 catalyzed (-)verbenone 10-hydroxylation at Vmax values of 15 and 21 nmol/min/nmol p450 with apparent Km values of 16 and 91 microM, respectively. verbenone 46-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 12867494-7 2003 In the rat, recombinant CYP2C11, CYP2B1, and CYP3A2 catalyzed (-)-verbenone 10-hydroxylation with Vmax and Km ratios (ml/min/nmol p450) of 0.73, 0.20, and 0.03, respectively. verbenone 62-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-134 12844133-0 2003 Artemisinin autoinduction is caused by involvement of cytochrome P450 2B6 but not 2C9. artemisinin 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-73 12844133-1 2003 AIM: Our goal was to investigate whether artemisinin autoinduction is caused by an increase in cytochrome P450 (CYP) 2B6 or CYP2C9 activities, we evaluated the effects of multiple-dose artemisinin administration on S-mephenytoin N-demethylation in healthy subjects. artemisinin 41-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-120 12844133-8 2003 The area under the concentration-time curve ratio of S-nirvanol/S-mephenytoin, an index of CYP2B6 activity, increased 1.9-fold (P <.05) in CYP2C19 poor metabolizers during artemisinin multiple-dose administration, whereas the urinary excretion ratio of hydroxytolbutamide plus carboxytolbutamide/tolbutamide remained constant during the study period. ethylphenylhydantoin 53-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 12844133-8 2003 The area under the concentration-time curve ratio of S-nirvanol/S-mephenytoin, an index of CYP2B6 activity, increased 1.9-fold (P <.05) in CYP2C19 poor metabolizers during artemisinin multiple-dose administration, whereas the urinary excretion ratio of hydroxytolbutamide plus carboxytolbutamide/tolbutamide remained constant during the study period. Mephenytoin 64-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 12844133-9 2003 CONCLUSIONS: These results indicate that artemisinin induces the N-demethylation of S-mephenytoin probably by an increased capacity of CYP2B6. artemisinin 41-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 12844133-9 2003 CONCLUSIONS: These results indicate that artemisinin induces the N-demethylation of S-mephenytoin probably by an increased capacity of CYP2B6. Nitrogen 2-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 12844133-9 2003 CONCLUSIONS: These results indicate that artemisinin induces the N-demethylation of S-mephenytoin probably by an increased capacity of CYP2B6. Mephenytoin 84-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 12844133-10 2003 The autoinduction phenomenon of artemisinin may, therefore, be attributed, at least in part, to induction of CYP2B6, because this is the isozyme primarily involved in its metabolism. artemisinin 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. NADP 86-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-32 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. NADP 86-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. NADP 86-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. Carbon Monoxide 118-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-32 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. Carbon Monoxide 118-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. Carbon Monoxide 118-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. Proadifen 186-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-32 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. Proadifen 186-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. Proadifen 186-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. Proadifen 211-219 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-32 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. Proadifen 211-219 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. Proadifen 211-219 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. 1-aminobenzotriazole 225-245 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-32 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. 1-aminobenzotriazole 225-245 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. 1-aminobenzotriazole 225-245 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. 1-aminobenzotriazole 247-250 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-32 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. 1-aminobenzotriazole 247-250 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12920169-7 2003 Contributions of cytochrome p450 (p450) enzymes to NNK oxidation were demonstrated by NADPH dependence, inhibition by carbon monoxide, and inhibition by the nonselective p450 inhibitors proadifen hydrochloride (SKF-525A) and 1-aminobenzotriazole (ABT), particularly in lung microsomes from high bioactivators. 1-aminobenzotriazole 247-250 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 12872138-0 2003 Sustained P450 expression and prodrug activation in bolus cyclophosphamide-treated cultured tumor cells. Cyclophosphamide 58-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-14 12872138-2 2003 Cytochrome P450-based gene therapy can substantially increase the sensitivity of tumor cells to P450-activated cancer chemotherapeutic prodrugs such as cyclophosphamide (CPA) without increasing host toxicity. Cyclophosphamide 152-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 12872138-2 2003 Cytochrome P450-based gene therapy can substantially increase the sensitivity of tumor cells to P450-activated cancer chemotherapeutic prodrugs such as cyclophosphamide (CPA) without increasing host toxicity. Cyclophosphamide 152-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-100 12872138-2 2003 Cytochrome P450-based gene therapy can substantially increase the sensitivity of tumor cells to P450-activated cancer chemotherapeutic prodrugs such as cyclophosphamide (CPA) without increasing host toxicity. Cyclophosphamide 170-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 12872138-2 2003 Cytochrome P450-based gene therapy can substantially increase the sensitivity of tumor cells to P450-activated cancer chemotherapeutic prodrugs such as cyclophosphamide (CPA) without increasing host toxicity. Cyclophosphamide 170-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-100 12872138-3 2003 While the role of 4-OH-CPA, the primary active metabolite of CPA, in eliciting tumor cell death is well established, the effect of 4-OH-CPA exposure on the capacity of P450-expressing tumor cells for continued metabolism and activation of CPA has not been investigated. 4-oh-cpa 131-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 168-172 12872138-4 2003 The present study addresses this question and characterizes the impact of CPA dose and treatment schedule on the ability of P450-expressing tumor cells to sustain prodrug activation over time. Cyclophosphamide 74-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 12872138-5 2003 9L gliosarcoma cells expressing human P450 2B6 and treated with CPA in a continuous manner exhibited a time- and CPA dose-dependent decrease in P450-catalyzed CPA 4-hydroxylase activity. Cyclophosphamide 64-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 12872138-5 2003 9L gliosarcoma cells expressing human P450 2B6 and treated with CPA in a continuous manner exhibited a time- and CPA dose-dependent decrease in P450-catalyzed CPA 4-hydroxylase activity. Cyclophosphamide 64-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-148 12872138-5 2003 9L gliosarcoma cells expressing human P450 2B6 and treated with CPA in a continuous manner exhibited a time- and CPA dose-dependent decrease in P450-catalyzed CPA 4-hydroxylase activity. Cyclophosphamide 113-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 12872138-5 2003 9L gliosarcoma cells expressing human P450 2B6 and treated with CPA in a continuous manner exhibited a time- and CPA dose-dependent decrease in P450-catalyzed CPA 4-hydroxylase activity. Cyclophosphamide 113-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-148 12872138-6 2003 This decrease reflects a selective, 4-OH-CPA-induced loss of cellular P450 protein content. 4-oh-cpa 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 12872138-7 2003 By contrast, when the P450-expressing tumor cells were treated with CPA as a single 8 hours exposure, cellular CPA 4-hydroxylase activity and P450 protein expression were substantially prolonged when compared to continuous prodrug treatment. Cyclophosphamide 68-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 12872138-7 2003 By contrast, when the P450-expressing tumor cells were treated with CPA as a single 8 hours exposure, cellular CPA 4-hydroxylase activity and P450 protein expression were substantially prolonged when compared to continuous prodrug treatment. Cyclophosphamide 68-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 12872138-8 2003 This schedule-dependent effect of CPA was influenced by the level of P450 protein expressed in the tumor cells. Cyclophosphamide 34-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-73 12872138-9 2003 At high P450 protein and activity levels, which could be achieved by culturing the tumor cells at high cell density, net production and release of 4-OH-CPA into the culture media was increased substantially. 4-oh-cpa 147-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-12 12872138-11 2003 These findings demonstrate the impact of CPA dose and treatment schedule on the efficacy of P450 gene-directed enzyme prodrug therapy, with bolus CPA treatment being compatible with sustained expression of P450 protein and maintenance of P450-dependent prodrug activation by the target tumor tissue. Cyclophosphamide 41-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 12872138-11 2003 These findings demonstrate the impact of CPA dose and treatment schedule on the efficacy of P450 gene-directed enzyme prodrug therapy, with bolus CPA treatment being compatible with sustained expression of P450 protein and maintenance of P450-dependent prodrug activation by the target tumor tissue. Cyclophosphamide 146-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 12872138-11 2003 These findings demonstrate the impact of CPA dose and treatment schedule on the efficacy of P450 gene-directed enzyme prodrug therapy, with bolus CPA treatment being compatible with sustained expression of P450 protein and maintenance of P450-dependent prodrug activation by the target tumor tissue. Cyclophosphamide 146-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 206-210 12872138-11 2003 These findings demonstrate the impact of CPA dose and treatment schedule on the efficacy of P450 gene-directed enzyme prodrug therapy, with bolus CPA treatment being compatible with sustained expression of P450 protein and maintenance of P450-dependent prodrug activation by the target tumor tissue. Cyclophosphamide 146-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 206-210 12818727-5 2003 Ethoxyresorufin, coumarin, benzoxyresorufin, chlorzoxazone, testosterone and lauric acid were used as selective substrates for CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2B6, CYP2E1, CYP3A4 and CYP4A, respectively. ethoxyresorufin 0-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 12818727-5 2003 Ethoxyresorufin, coumarin, benzoxyresorufin, chlorzoxazone, testosterone and lauric acid were used as selective substrates for CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2B6, CYP2E1, CYP3A4 and CYP4A, respectively. lauric acid 77-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 12814972-0 2003 In vitro metabolism of midazolam, triazolam, nifedipine, and testosterone by human liver microsomes and recombinant cytochromes p450: role of cyp3a4 and cyp3a5. Testosterone 61-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 12814665-1 2003 CYP2B6 metabolizes drugs such as nicotine and bupropion, and many toxins and carcinogens. Bupropion 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 12814665-2 2003 Nicotine induces CYP2B1 in rat brain and in humans polymorphic variation in CYP2B6 affects smoking cessation rates. Nicotine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 12814665-9 2003 Higher brain CYP2B6 activity in smokers and alcoholics may cause altered sensitivity to centrally acting drugs, increased susceptibility to neurotoxins and carcinogenic xenobiotics and contribute to central tolerance to nicotine. Nicotine 220-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 12695351-4 2003 In contrast, treatment with 0.1 microM DEX enhanced CYP2B6 induction by different pregnane X receptor (PXR) activators, including rifampin, phenytoin, clotrimazole, and phenobarbital. Dexamethasone 39-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 12676886-4 2003 In a panel of 10 P450s, CYP2B6 formed 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) at the highest rate. 8-hydroxyefavirenz 38-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 12676886-4 2003 In a panel of 10 P450s, CYP2B6 formed 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) at the highest rate. 8,14-Dihydroxyefavirenz 61-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 12676886-5 2003 The Km value for the formation of 8-hydroxyefavirenz in CYP2B6 derived from hyperbolic Eq. 12.4 micro M) was close to that obtained in HLMs (Km, 20.2 micro M). 8-hydroxyefavirenz 34-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 12676886-7 2003 When 8-hydroxyefavirenz (2.5 micro M) was used as a substrate, 8,14-dihydroxyefavirenz was formed by CYP2B6 at the highest rate, and its kinetics showed substrate inhibition (Ksi, approximately 94 micro M in HLMs and approximately 234 micro M in CYP2B6). 8-hydroxyefavirenz 5-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 12676886-7 2003 When 8-hydroxyefavirenz (2.5 micro M) was used as a substrate, 8,14-dihydroxyefavirenz was formed by CYP2B6 at the highest rate, and its kinetics showed substrate inhibition (Ksi, approximately 94 micro M in HLMs and approximately 234 micro M in CYP2B6). 8-hydroxyefavirenz 5-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 246-252 12676886-7 2003 When 8-hydroxyefavirenz (2.5 micro M) was used as a substrate, 8,14-dihydroxyefavirenz was formed by CYP2B6 at the highest rate, and its kinetics showed substrate inhibition (Ksi, approximately 94 micro M in HLMs and approximately 234 micro M in CYP2B6). 8,14-Dihydroxyefavirenz 63-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 12676886-7 2003 When 8-hydroxyefavirenz (2.5 micro M) was used as a substrate, 8,14-dihydroxyefavirenz was formed by CYP2B6 at the highest rate, and its kinetics showed substrate inhibition (Ksi, approximately 94 micro M in HLMs and approximately 234 micro M in CYP2B6). 8,14-Dihydroxyefavirenz 63-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 246-252 12676886-7 2003 When 8-hydroxyefavirenz (2.5 micro M) was used as a substrate, 8,14-dihydroxyefavirenz was formed by CYP2B6 at the highest rate, and its kinetics showed substrate inhibition (Ksi, approximately 94 micro M in HLMs and approximately 234 micro M in CYP2B6). KS I 175-178 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 12676886-7 2003 When 8-hydroxyefavirenz (2.5 micro M) was used as a substrate, 8,14-dihydroxyefavirenz was formed by CYP2B6 at the highest rate, and its kinetics showed substrate inhibition (Ksi, approximately 94 micro M in HLMs and approximately 234 micro M in CYP2B6). KS I 175-178 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 246-252 12676886-8 2003 In a panel of 11 HLMs, 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz formation rates from efavirenz (10 micro M) correlated significantly with the activity of CYP2B6 and CYP3A. 8-hydroxyefavirenz 23-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 160-166 12676886-8 2003 In a panel of 11 HLMs, 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz formation rates from efavirenz (10 micro M) correlated significantly with the activity of CYP2B6 and CYP3A. 8,14-Dihydroxyefavirenz 46-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 160-166 12676886-9 2003 N,N",N"-Triethylenethiophosphoramide (thioTEPA; 50 micro M) inhibited the formation rates of 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) by > or = 60% in HLMs) and CYP2B6, with Ki values < 4 micro M. In conclusion, CYP2B6 is the principal catalyst of efavirenz sequential hydroxylation. Thiotepa 0-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 12676886-9 2003 N,N",N"-Triethylenethiophosphoramide (thioTEPA; 50 micro M) inhibited the formation rates of 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) by > or = 60% in HLMs) and CYP2B6, with Ki values < 4 micro M. In conclusion, CYP2B6 is the principal catalyst of efavirenz sequential hydroxylation. Thiotepa 0-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 252-258 12676886-9 2003 N,N",N"-Triethylenethiophosphoramide (thioTEPA; 50 micro M) inhibited the formation rates of 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) by > or = 60% in HLMs) and CYP2B6, with Ki values < 4 micro M. In conclusion, CYP2B6 is the principal catalyst of efavirenz sequential hydroxylation. Thiotepa 38-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 12676886-9 2003 N,N",N"-Triethylenethiophosphoramide (thioTEPA; 50 micro M) inhibited the formation rates of 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) by > or = 60% in HLMs) and CYP2B6, with Ki values < 4 micro M. In conclusion, CYP2B6 is the principal catalyst of efavirenz sequential hydroxylation. Thiotepa 38-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 252-258 12676886-9 2003 N,N",N"-Triethylenethiophosphoramide (thioTEPA; 50 micro M) inhibited the formation rates of 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) by > or = 60% in HLMs) and CYP2B6, with Ki values < 4 micro M. In conclusion, CYP2B6 is the principal catalyst of efavirenz sequential hydroxylation. 8-hydroxyefavirenz 93-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 12676886-9 2003 N,N",N"-Triethylenethiophosphoramide (thioTEPA; 50 micro M) inhibited the formation rates of 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) by > or = 60% in HLMs) and CYP2B6, with Ki values < 4 micro M. In conclusion, CYP2B6 is the principal catalyst of efavirenz sequential hydroxylation. 8-hydroxyefavirenz 93-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 252-258 12676886-9 2003 N,N",N"-Triethylenethiophosphoramide (thioTEPA; 50 micro M) inhibited the formation rates of 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) by > or = 60% in HLMs) and CYP2B6, with Ki values < 4 micro M. In conclusion, CYP2B6 is the principal catalyst of efavirenz sequential hydroxylation. 8,14-Dihydroxyefavirenz 116-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 12676886-9 2003 N,N",N"-Triethylenethiophosphoramide (thioTEPA; 50 micro M) inhibited the formation rates of 8-hydroxyefavirenz and 8,14-dihydroxyefavirenz from efavirenz (10 micro M) by > or = 60% in HLMs) and CYP2B6, with Ki values < 4 micro M. In conclusion, CYP2B6 is the principal catalyst of efavirenz sequential hydroxylation. 8,14-Dihydroxyefavirenz 116-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 252-258 12756210-0 2003 Interaction of buprenorphine and its metabolite norbuprenorphine with cytochromes p450 in vitro. Buprenorphine 15-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 12756210-0 2003 Interaction of buprenorphine and its metabolite norbuprenorphine with cytochromes p450 in vitro. norbuprenorphine 48-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 12756210-2 2003 In this study, the selective probe reactions for each of the major hepatic cytochromes P450 (P450s) were used to evaluate the effect of buprenorphine and its main metabolite norbuprenorphine on the activity of these P450s. Buprenorphine 136-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 12756210-2 2003 In this study, the selective probe reactions for each of the major hepatic cytochromes P450 (P450s) were used to evaluate the effect of buprenorphine and its main metabolite norbuprenorphine on the activity of these P450s. Buprenorphine 136-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-98 12756210-2 2003 In this study, the selective probe reactions for each of the major hepatic cytochromes P450 (P450s) were used to evaluate the effect of buprenorphine and its main metabolite norbuprenorphine on the activity of these P450s. Buprenorphine 136-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 216-221 12756210-2 2003 In this study, the selective probe reactions for each of the major hepatic cytochromes P450 (P450s) were used to evaluate the effect of buprenorphine and its main metabolite norbuprenorphine on the activity of these P450s. norbuprenorphine 174-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 12756210-2 2003 In this study, the selective probe reactions for each of the major hepatic cytochromes P450 (P450s) were used to evaluate the effect of buprenorphine and its main metabolite norbuprenorphine on the activity of these P450s. norbuprenorphine 174-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-98 12756210-2 2003 In this study, the selective probe reactions for each of the major hepatic cytochromes P450 (P450s) were used to evaluate the effect of buprenorphine and its main metabolite norbuprenorphine on the activity of these P450s. norbuprenorphine 174-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 216-221 12756210-7 2003 The present in vitro study suggests that buprenorphine and its major metabolite norbuprenorphine are inhibitors of CYP2D6 and CYP3A4; however, at therapeutic concentrations they are not predicted to cause potentially clinically important drug interactions with other drugs metabolized by major hepatic P450s. Buprenorphine 41-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 302-307 12756210-7 2003 The present in vitro study suggests that buprenorphine and its major metabolite norbuprenorphine are inhibitors of CYP2D6 and CYP3A4; however, at therapeutic concentrations they are not predicted to cause potentially clinically important drug interactions with other drugs metabolized by major hepatic P450s. norbuprenorphine 80-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 302-307 12851038-14 2003 Since all the inhibition profiles of astemizole O-demethylation were different in the liver and small intestine, involvement of another p450 in astemizole O-demethylation in human liver may be speculated. Astemizole 144-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 12815172-4 2003 Colchicine (COL) decreased both basal and rifampicin- and phenobarbital-inducible expression of CYP2B6, CYP2C8/9, and CYP3A4. Colchicine 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 12815172-4 2003 Colchicine (COL) decreased both basal and rifampicin- and phenobarbital-inducible expression of CYP2B6, CYP2C8/9, and CYP3A4. Rifampin 42-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 12815172-4 2003 Colchicine (COL) decreased both basal and rifampicin- and phenobarbital-inducible expression of CYP2B6, CYP2C8/9, and CYP3A4. Phenobarbital 58-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 12893521-4 2003 7-Benzyloxyresorufin O-debenzylase activity in human liver microsomes was inhibited by a monoclonal antibody against CYP2B6 and a polyclonal antibody against CYP3A2 by 53-69 and 19-44%, respectively, suggesting that CYP2B6 and CYP3A4 mainly catalyse 7-benzyloxyresorufin O-debenzylation in human liver microsomes. benzyloxyresorufin 250-270 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 12893521-7 2003 Additionally, 7-benzyloxyresorufin O-debenzylation by recombinant CYP3A4 was increased by the addition of alpha-naphthoflavone, testosterone and progesterone but not by quinidine, whereas no chemicals tested could activate the O-debenzylation of 7-benzyloxyresorufin by CYP2B6. benzyloxyresorufin 14-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 270-276 12893521-7 2003 Additionally, 7-benzyloxyresorufin O-debenzylation by recombinant CYP3A4 was increased by the addition of alpha-naphthoflavone, testosterone and progesterone but not by quinidine, whereas no chemicals tested could activate the O-debenzylation of 7-benzyloxyresorufin by CYP2B6. alpha-naphthoflavone 106-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 270-276 12893521-7 2003 Additionally, 7-benzyloxyresorufin O-debenzylation by recombinant CYP3A4 was increased by the addition of alpha-naphthoflavone, testosterone and progesterone but not by quinidine, whereas no chemicals tested could activate the O-debenzylation of 7-benzyloxyresorufin by CYP2B6. Testosterone 128-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 270-276 12782579-14 2003 These results demonstrate for the first time the potential of human NADPH: p450 reductase and recombinant p450s to contribute to the metabolic activation of 3-NBA by nitroreduction. 3-nitrobenzanthrone 157-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 12870655-7 2003 Additional experiments showed that phenobarbital increased CYP2B6 mRNA expression and that pregnane X receptor (PXR) but not constitutive androstane receptor (CAR) was detected in HL-60 cells. Phenobarbital 35-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 12695351-4 2003 In contrast, treatment with 0.1 microM DEX enhanced CYP2B6 induction by different pregnane X receptor (PXR) activators, including rifampin, phenytoin, clotrimazole, and phenobarbital. Rifampin 130-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 12695351-4 2003 In contrast, treatment with 0.1 microM DEX enhanced CYP2B6 induction by different pregnane X receptor (PXR) activators, including rifampin, phenytoin, clotrimazole, and phenobarbital. Phenytoin 140-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 12695351-4 2003 In contrast, treatment with 0.1 microM DEX enhanced CYP2B6 induction by different pregnane X receptor (PXR) activators, including rifampin, phenytoin, clotrimazole, and phenobarbital. Clotrimazole 151-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 12695351-4 2003 In contrast, treatment with 0.1 microM DEX enhanced CYP2B6 induction by different pregnane X receptor (PXR) activators, including rifampin, phenytoin, clotrimazole, and phenobarbital. Phenobarbital 169-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 12695351-5 2003 In Huh7 cells, cotransfection of human (h)GR and hPXR with CYP2B6-phenobarbital-responsive enhancer module (PBREM) reporter constructs revealed that all hPXR ligands induce CYP2B6 reporter gene activity, and this ligand-dependent activation is greatly enhanced by activated hGR. Phenobarbital 66-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 12695351-8 2003 In the presence of activated hGR and known inducers of CYP2B6, only PB treatment caused a further 2-fold activation of hCAR compared with control. Lead 68-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 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 2 subfamily B member 6 Homo sapiens 11-15 12959413-5 2003 In contrast, the bulk of the evidence indicates that inactivated and/or otherwise posttranslationally modified P450 proteins undergo adenosine triphosphate-dependent proteolytic degradation in the cytosol. Adenosine Triphosphate 133-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-115 12721102-17 2003 The highest intrinsic clearance (V(max)/K(m)) was found for CYP1A subfamily, CYP3A4 and CYP2B6 in the case of 5- sulphoxidation, and for CYP2C19, CYP1A subfamily and CYP2B6 in the case of N-demethylation. Nitrogen 188-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 12571232-1 2003 CYP2B6 plays an important role in the metabolism of a variety of structurally unrelated xenobiotics, including the anticancer drugs cyclophosphamide and ifosfamide. Cyclophosphamide 132-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 12571232-1 2003 CYP2B6 plays an important role in the metabolism of a variety of structurally unrelated xenobiotics, including the anticancer drugs cyclophosphamide and ifosfamide. Ifosfamide 153-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 12721102-9 2003 promazine metabolism in a primary culture of human hepatocytes treated with specific inducers (rifampicin-CYP3A4, CYP2B6 and CYP2C inducer, 2,3,7,8-tetrachlordibenzeno-p-dioxin (TCDD)-CYP1A1/1A2 inducer). Promazine 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 12646859-6 2003 Next, we showed that an adenovirus expressing the human cytochrome P450 (CYP2B6) regulated by the OBHRE promoter delays tumour growth in response to the prodrug cyclophosphamide (CPA). Cyclophosphamide 161-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 12642463-1 2003 Cytochrome P450 (P450) enzymes are major catalysts involved in the metabolism of xenobiotics and endogenous substrates such as testosterone (TST). Testosterone 127-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 12642463-1 2003 Cytochrome P450 (P450) enzymes are major catalysts involved in the metabolism of xenobiotics and endogenous substrates such as testosterone (TST). Testosterone 141-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 12732316-2 2003 Engineered P450 expression is needed for low-cost production of antineoplastic drugs such as taxol or indole alkaloids and offers the possibility to increase the content of nutraceuticals such as phytoestrogens and antioxidants in plants. Paclitaxel 93-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 12732316-2 2003 Engineered P450 expression is needed for low-cost production of antineoplastic drugs such as taxol or indole alkaloids and offers the possibility to increase the content of nutraceuticals such as phytoestrogens and antioxidants in plants. Indole Alkaloids 102-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 12642470-0 2003 Comparative effects of thiazolidinediones on in vitro P450 enzyme induction and inhibition. Thiazolidinediones 23-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 12646859-6 2003 Next, we showed that an adenovirus expressing the human cytochrome P450 (CYP2B6) regulated by the OBHRE promoter delays tumour growth in response to the prodrug cyclophosphamide (CPA). Cyclophosphamide 179-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 12619052-3 2003 The metabolic fate of (14)C-artemisinin in microsomes from human B-lymphoblastoid cell lines transformed with CYP2A6, CYP2B6 and CYP3A4 was also investigated. artemisinin 28-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 12642470-5 2003 A comparable concentration-dependent increase in CYP2B6 immunoreactive protein was observed with all three thiazolidinediones. Thiazolidinediones 107-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 12642470-11 2003 This is the first report of in vitro induction of P450 enzymes by rosiglitazone and pioglitazone. Rosiglitazone 66-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 12642470-11 2003 This is the first report of in vitro induction of P450 enzymes by rosiglitazone and pioglitazone. Pioglitazone 84-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 12643683-0 2003 Epoxidation of olefins by hydroperoxo-ferric cytochrome P450. Alkenes 15-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 12643683-4 2003 The present results provide strong evidence that hydroperoxo-ferric P450 can serve as a second electrophilic oxidant capable of olefin epoxidation. Alkenes 128-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 12739762-0 2003 Induction of cytochrome P450 2B6 and 3A4 expression by phenobarbital and cyclophosphamide in cultured human liver slices. Phenobarbital 55-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-32 12739762-0 2003 Induction of cytochrome P450 2B6 and 3A4 expression by phenobarbital and cyclophosphamide in cultured human liver slices. Cyclophosphamide 73-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-32 12739762-8 2003 CYP2B6 and 3A4 mRNA, apoprotein, and enzyme-related activities were induced by phenobarbital and cyclophosphamide, whereas CYP2C9 apoprotein was not. Phenobarbital 79-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 12739762-8 2003 CYP2B6 and 3A4 mRNA, apoprotein, and enzyme-related activities were induced by phenobarbital and cyclophosphamide, whereas CYP2C9 apoprotein was not. Cyclophosphamide 97-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 12527701-0 2003 Comparison of the formation of N-alkylprotoporphyrin IX after interaction of porphyrinogenic xenobiotics with single cDNA-expressed human P450 enzymes in microsomes prepared from baculovirus-infected insect cells and human lymphoblastoid cell lines. n-alkylprotoporphyrin ix 31-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-142 12598790-0 2003 Concurrent use of bupropion with CYP2B6 inhibitors, nelfinavir, ritonavir and efavirenz: a case series. Bupropion 18-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 12527701-2 2003 In this study, we tested the hypothesis that N-alkylprotoporphyrin IX (N-alkylPP) formation following interaction of porphyrinogenic xenobiotics with single cDNA-expressed human P450 enzymes in microsomes from HLCL would occur only with P450 enzymes that had undergone mechanism-based inactivation. n-alkylprotoporphyrin ix 45-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 12527701-2 2003 In this study, we tested the hypothesis that N-alkylprotoporphyrin IX (N-alkylPP) formation following interaction of porphyrinogenic xenobiotics with single cDNA-expressed human P450 enzymes in microsomes from HLCL would occur only with P450 enzymes that had undergone mechanism-based inactivation. n-alkylprotoporphyrin ix 45-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 237-241 12527701-2 2003 In this study, we tested the hypothesis that N-alkylprotoporphyrin IX (N-alkylPP) formation following interaction of porphyrinogenic xenobiotics with single cDNA-expressed human P450 enzymes in microsomes from HLCL would occur only with P450 enzymes that had undergone mechanism-based inactivation. n-alkylpp 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 12527701-2 2003 In this study, we tested the hypothesis that N-alkylprotoporphyrin IX (N-alkylPP) formation following interaction of porphyrinogenic xenobiotics with single cDNA-expressed human P450 enzymes in microsomes from HLCL would occur only with P450 enzymes that had undergone mechanism-based inactivation. n-alkylpp 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 237-241 12527701-5 2003 Fluorometry was used to measure N-alkylPP formation following interaction of porphyrinogenic xenobiotics and NADPH with cDNA-expressed human P450 enzymes in microsomes from HLCL or BIICL. Nitrogen 32-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 12527701-5 2003 Fluorometry was used to measure N-alkylPP formation following interaction of porphyrinogenic xenobiotics and NADPH with cDNA-expressed human P450 enzymes in microsomes from HLCL or BIICL. NADP 109-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 12527701-5 2003 Fluorometry was used to measure N-alkylPP formation following interaction of porphyrinogenic xenobiotics and NADPH with cDNA-expressed human P450 enzymes in microsomes from HLCL or BIICL. biicl 181-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 12527701-6 2003 With TTMS and 4-ethylDDC but not with AIA, N-alkylPP formation was observed only with human P450 enzymes CYP2D6, 1A2, 3A4, or 2C9 in microsomes from HLCL, which had undergone mechanism-based inactivation. 3-(2-(2,4,6-trimethylphenyl)thioethyl)-4-methylsydnone 5-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-96 12464245-8 2003 The epoxides and other products provide a view of the landscape of P450-generated reactive products and the myriad of chemistry involved in the metabolism of drugs and protoxicants. Epoxy Compounds 4-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-71 12485953-1 2003 The prodrug clopidogrel (Plavix) is activated by cytochrome p450 (p450) to a metabolite that inhibits ADP-induced platelet aggregation. Clopidogrel 25-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 12464242-2 2003 From a consideration of specific interactions between drug substrates for human CYP2 family enzymes and the putative active sites of CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP2E1, it is likely that the number and disposition of hydrogen bond donor/acceptors and aromatic rings within the various P450 substrate molecules determines their enzyme selectivity and binding affinity, together with directing their preferred routes of metabolism by the CYP2 enzymes concerned. Hydrogen 242-250 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 12485950-0 2003 Mechanism-based inactivation of cytochrome P450 2B6 by a novel terminal acetylene inhibitor. Acetylene 72-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-51 12485953-1 2003 The prodrug clopidogrel (Plavix) is activated by cytochrome p450 (p450) to a metabolite that inhibits ADP-induced platelet aggregation. Adenosine Diphosphate 102-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-64 12485953-1 2003 The prodrug clopidogrel (Plavix) is activated by cytochrome p450 (p450) to a metabolite that inhibits ADP-induced platelet aggregation. Adenosine Diphosphate 102-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 12485950-1 2003 N-(3,5-Dichloro-4-pyridyl)-3-(cyclopentyloxy)-4-methoxybenzamide (DCMB) is a known marker substrate for cytochrome p450 2B6. n-(3,5-dichloro-4-pyridyl)-3-(cyclopentyloxy)-4-methoxybenzamide 0-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-123 12485953-4 2003 Microsomes prepared from dexamethasone-pretreated rats metabolized clopidogrel at a rate of 3.8 nmol min(-1) nmol of p450(-1), which is 65 and 1270% faster than the rate of metabolism by microsomes from control and beta-napthoflavone-treated rats, respectively. Dexamethasone 25-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-121 12485950-1 2003 N-(3,5-Dichloro-4-pyridyl)-3-(cyclopentyloxy)-4-methoxybenzamide (DCMB) is a known marker substrate for cytochrome p450 2B6. DCMB 66-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-123 12485945-1 2003 CYP3A4, a cytochrome p450 (p450) isoform metabolizes estrogens, whereas CYP3A7, a fetal liver p450 isoform, is involved in estriol biosynthesis. Estriol 123-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 12485945-1 2003 CYP3A4, a cytochrome p450 (p450) isoform metabolizes estrogens, whereas CYP3A7, a fetal liver p450 isoform, is involved in estriol biosynthesis. Estriol 123-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 12485953-4 2003 Microsomes prepared from dexamethasone-pretreated rats metabolized clopidogrel at a rate of 3.8 nmol min(-1) nmol of p450(-1), which is 65 and 1270% faster than the rate of metabolism by microsomes from control and beta-napthoflavone-treated rats, respectively. Clopidogrel 67-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-121 12485945-1 2003 CYP3A4, a cytochrome p450 (p450) isoform metabolizes estrogens, whereas CYP3A7, a fetal liver p450 isoform, is involved in estriol biosynthesis. Estriol 123-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 12485952-0 2003 The mechanism-based inactivation of human cytochrome P450 2B6 by phencyclidine. Phencyclidine 65-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-61 12485953-4 2003 Microsomes prepared from dexamethasone-pretreated rats metabolized clopidogrel at a rate of 3.8 nmol min(-1) nmol of p450(-1), which is 65 and 1270% faster than the rate of metabolism by microsomes from control and beta-napthoflavone-treated rats, respectively. beta-napthoflavone 215-233 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-121 12485952-1 2003 Phencyclidine (PCP) was analyzed for its ability to inactivate human cytochrome p450 (p450) 2B6. Phencyclidine 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 12485952-1 2003 Phencyclidine (PCP) was analyzed for its ability to inactivate human cytochrome p450 (p450) 2B6. Phencyclidine 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-90 12485952-1 2003 Phencyclidine (PCP) was analyzed for its ability to inactivate human cytochrome p450 (p450) 2B6. Phencyclidine 15-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 12485953-5 2003 To identify the human p450s responsible for clopidogrel oxidation, genetically engineered microsomes containing a single human p450 isozyme were tested for their ability to oxidize clopidogrel. Clopidogrel 44-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 12485952-1 2003 Phencyclidine (PCP) was analyzed for its ability to inactivate human cytochrome p450 (p450) 2B6. Phencyclidine 15-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-90 12485953-6 2003 CYP3A4 and 3A5 metabolized clopidogrel at a significantly higher rate than eight other p450 isozymes, suggesting that CYP3A4 and 3A5 are primarily responsible for in vivo clopidogrel metabolism. Clopidogrel 171-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 12485952-2 2003 PCP inactivated the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation activity of p450 2B6 in a concentration-, time-, and NADPH-dependent manner and exhibited pseudo-first order kinetics. Phencyclidine 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 12485953-7 2003 Clopidogrel interacts with human CYP3A4 with a spectral dissociation constant (K(s)), K(m), and V(max) of 12 microM, 14 +/- 1 microM and 6.7 +/- 1 nmol min(-1) nmol p450(-1), respectively. Clopidogrel 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-169 12485952-2 2003 PCP inactivated the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation activity of p450 2B6 in a concentration-, time-, and NADPH-dependent manner and exhibited pseudo-first order kinetics. 7-ethoxy-4-trifluoromethylcoumarin 20-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 12485953-10 2003 Since CYP3A4 and 3A5 metabolize clopidogrel faster than other human p450 isozymes and are the most abundant p450s in human liver, they are predicted to be predominantly responsible for the activation of clopidogrel in vivo. Clopidogrel 203-214 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 12485952-2 2003 PCP inactivated the 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation activity of p450 2B6 in a concentration-, time-, and NADPH-dependent manner and exhibited pseudo-first order kinetics. NADP 125-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 12485952-6 2003 Extensive dialysis of the PCP-inactivated p450 2B6 did not cause a return in catalytic activity demonstrating PCP inactivation was irreversible. Phencyclidine 26-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 12485954-0 2003 Differences in cytochrome P450 forms involved in the metabolism of N,N-dipropyl-2-[4-methoxy-3-(2-phenylethoxy)phenyl]ethylamine monohydrochloride (NE-100), a novel sigma ligand, in human liver and intestine. N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 67-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 12485952-9 2003 High performance liquid chromatography analysis of p450 2B6 inactivated in the presence of (3)H-labeled PCP showed that PCP binding was specific for the p450 and not to other proteins in the reaction mixture. Tritium 91-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 12485952-9 2003 High performance liquid chromatography analysis of p450 2B6 inactivated in the presence of (3)H-labeled PCP showed that PCP binding was specific for the p450 and not to other proteins in the reaction mixture. Tritium 91-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 12485952-9 2003 High performance liquid chromatography analysis of p450 2B6 inactivated in the presence of (3)H-labeled PCP showed that PCP binding was specific for the p450 and not to other proteins in the reaction mixture. Phencyclidine 104-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 12485952-9 2003 High performance liquid chromatography analysis of p450 2B6 inactivated in the presence of (3)H-labeled PCP showed that PCP binding was specific for the p450 and not to other proteins in the reaction mixture. Phencyclidine 104-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 12485952-9 2003 High performance liquid chromatography analysis of p450 2B6 inactivated in the presence of (3)H-labeled PCP showed that PCP binding was specific for the p450 and not to other proteins in the reaction mixture. Phencyclidine 120-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 12485952-9 2003 High performance liquid chromatography analysis of p450 2B6 inactivated in the presence of (3)H-labeled PCP showed that PCP binding was specific for the p450 and not to other proteins in the reaction mixture. Phencyclidine 120-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 12485952-10 2003 The stoichiometry of binding of PCP to p450 2B6 was demonstrated using (3)H-labeled PCP. Phencyclidine 32-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 12485952-10 2003 The stoichiometry of binding of PCP to p450 2B6 was demonstrated using (3)H-labeled PCP. Phencyclidine 84-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 12485954-0 2003 Differences in cytochrome P450 forms involved in the metabolism of N,N-dipropyl-2-[4-methoxy-3-(2-phenylethoxy)phenyl]ethylamine monohydrochloride (NE-100), a novel sigma ligand, in human liver and intestine. N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 148-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 12485953-0 2003 The metabolism of clopidogrel is catalyzed by human cytochrome P450 3A and is inhibited by atorvastatin. Clopidogrel 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 12485954-7 2003 The activity of NE-100 metabolism was inhibited by approximately 80% by an anti-CYP2D6 antibody and only by quinidine among the p450-selective inhibitors at a low substrate concentration (0.1 microM). N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 16-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 12485953-1 2003 The prodrug clopidogrel (Plavix) is activated by cytochrome p450 (p450) to a metabolite that inhibits ADP-induced platelet aggregation. Clopidogrel 12-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-64 12490624-7 2003 Substitution of residue 363 in CYP2E1 and CYP2B6 resulted in significant alterations of the metabolite profile for the side chain hydroxylation of 7-butoxycoumarin. 7-butoxy-2H-chromen-2-one 147-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 12485953-1 2003 The prodrug clopidogrel (Plavix) is activated by cytochrome p450 (p450) to a metabolite that inhibits ADP-induced platelet aggregation. Clopidogrel 12-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 12485953-1 2003 The prodrug clopidogrel (Plavix) is activated by cytochrome p450 (p450) to a metabolite that inhibits ADP-induced platelet aggregation. Clopidogrel 25-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-64 12517592-3 2003 Total RNA was examined by Northern blot analysis using (32)P-labeled cDNA probes encoding human P450 side chain cleavage (P450(SSC)), P450(17alpha), and cytochrome P450 aromatase (P450(arom)). Phosphorus-32 55-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-100 12824576-0 2003 High-throughput carbon monoxide binding assay for cytochromes p450. Carbon Monoxide 16-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 12485961-5 2003 The formation of the benzoquinone imine [detected as a glutathione (GSH) adduct, M5] was primarily carried out by CYP3A4, whereas M8 was formed mainly by the polymorphic CYP2B6. p-Benzoquinone imine 21-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-176 12485961-11 2003 These results suggest that CYP2B6 has a higher affinity than CYP3A4 toward DPC 963. dpc 75-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 12485961-12 2003 This consequently leads to greater levels of CYP2B6-catalyzed product, M8, than CYP3A4-mediated bioactivation of DPC 963 to benzoquinone imine or oxirene intermediates. dpc 113-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 12490624-4 2003 One of the most intriguing findings was that substitution of CYP2E1 Phe-477 with valine from CYP2B6 resulted in significant 7-ethoxy-4-trifluoromethylcoumarin deethylation. 7-ethoxy-4-trifluoromethylcoumarin 124-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 12490624-5 2003 Use of three-dimensional models of CYP2B6 and CYP2E1 based on the crystal structure of CYP2C5 suggested that deethylation of 7-ethoxy-4-trifluoromethylcoumarin by CYP2E1 is impeded by van der Waals overlaps with the side chain of Phe-477. 7-ethoxy-4-trifluoromethylcoumarin 125-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 12490624-5 2003 Use of three-dimensional models of CYP2B6 and CYP2E1 based on the crystal structure of CYP2C5 suggested that deethylation of 7-ethoxy-4-trifluoromethylcoumarin by CYP2E1 is impeded by van der Waals overlaps with the side chain of Phe-477. Phenylalanine 230-233 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 12629583-0 2003 Role of polymorphic human CYP2B6 in cyclophosphamide bioactivation. Cyclophosphamide 36-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 12556907-1 2003 In recent studies that addressed the transcriptional control of steroid synthesis, a transcriptional regulating protein of 132 kDa (TReP-132) was cloned and demonstrated to regulate expression of the human P450 side chain cleavage (P450scc) gene. Steroids 64-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 206-210 12629583-1 2003 The role of polymorphic CYP2B6 in cyclophosphamide (CPA) bioactivation was investigated in human liver microsomes. Cyclophosphamide 52-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 12511605-1 2003 The constitutive androstane receptor (CAR, NR1I3) transcriptionally activates cytochrome P450 2B6, 2C9, and 3A4 when activated by xenobiotics, such as phenobarbital. Phenobarbital 151-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-102 12629583-1 2003 The role of polymorphic CYP2B6 in cyclophosphamide (CPA) bioactivation was investigated in human liver microsomes. Cyclophosphamide 34-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 12629583-13 2003 Our results demonstrate that the polymorphic CYP2B6 is a major enzyme in the bioactivation of CPA. Cyclophosphamide 94-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 12519691-10 2003 However, other cytochrome P450 enzymes (CYP2E1 and CYP2B6) also appear to play a role in the N-oxidation of this drug. Nitrogen 93-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 12433799-11 2002 In conclusion, enantiomeric abundance of methoxychlor metabolites depends on the relative catalytic activity of the hepatic p450 isoforms. Methoxychlor 41-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 12433806-0 2002 Multiple cytochrome P450 enzymes responsible for the oxidative metabolism of the substituted (S)-3-phenylpiperidine, (S,S)-3-[3-(methylsulfonyl)phenyl]-1-propylpiperidine hydrochloride, in human liver microsomes. (S)-3-Phenylpiperidine 93-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 12460909-1 2002 Cytochrome P450 gene-directed enzyme prodrug therapy substantially augments intratumoral activation of anticancer prodrugs, such as cyclophosphamide (CPA), leading to a strong increase in antitumor effect without a corresponding increase in host toxicity. Cyclophosphamide 132-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 12460909-1 2002 Cytochrome P450 gene-directed enzyme prodrug therapy substantially augments intratumoral activation of anticancer prodrugs, such as cyclophosphamide (CPA), leading to a strong increase in antitumor effect without a corresponding increase in host toxicity. Cyclophosphamide 150-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 12460909-2 2002 Attempts to additionally increase tumor cell kill by enhancing the intrinsic chemosensitivity of P450-expressing tumor cells by chemical means (depletion of cellular glutathione) or by coexpression of proapoptotic factors was shown to result in the desired increase in chemosensitivity, but with a decrease in net production of bystander cytotoxic drug metabolites because of accelerated death of the prodrug-activating tumor cells. Glutathione 166-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-101 12460909-3 2002 Moreover, tumor cell P450 activity declined during the course of apoptosis induced by P450-activated CPA, limiting the potential of the tumor cell for continued production of activated drug metabolites. Cyclophosphamide 101-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 12460909-3 2002 Moreover, tumor cell P450 activity declined during the course of apoptosis induced by P450-activated CPA, limiting the potential of the tumor cell for continued production of activated drug metabolites. Cyclophosphamide 101-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-90 12433799-0 2002 Enantioselective metabolism of the endocrine disruptor pesticide methoxychlor by human cytochromes P450 (P450s): major differences in selective enantiomer formation by various P450 isoforms. Methoxychlor 65-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-103 12433799-0 2002 Enantioselective metabolism of the endocrine disruptor pesticide methoxychlor by human cytochromes P450 (P450s): major differences in selective enantiomer formation by various P450 isoforms. Methoxychlor 65-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-109 12433799-4 2002 Since methoxychlor-metabolite enantiomers may have different estrogenic/antiestrogenic/antiandrogenic activities than corresponding racemates, the possibility that p450s preferentially generate or use R or S enantiomers, was examined. Methoxychlor 6-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-168 12433806-8 2002 In addition, the selectivity of (-)-OSU6162 to inhibit six human p450 enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2E1, CYP2D6 and CYP3A4) was evaluated using an in vitro inhibition screen. OSU 6162 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 12433806-0 2002 Multiple cytochrome P450 enzymes responsible for the oxidative metabolism of the substituted (S)-3-phenylpiperidine, (S,S)-3-[3-(methylsulfonyl)phenyl]-1-propylpiperidine hydrochloride, in human liver microsomes. (s,s)-3-[3-(methylsulfonyl)phenyl]-1-propylpiperidine hydrochloride 117-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 12433806-3 2002 Kinetics evidence is presented that the N-depropylation of (-)-OSU6162 in human hepatic microsomes is mediated by multiple cytochrome p450 (p450) enzymes, in particular CYP2D6. Nitrogen 40-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-138 12433806-3 2002 Kinetics evidence is presented that the N-depropylation of (-)-OSU6162 in human hepatic microsomes is mediated by multiple cytochrome p450 (p450) enzymes, in particular CYP2D6. Nitrogen 40-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-144 12433806-3 2002 Kinetics evidence is presented that the N-depropylation of (-)-OSU6162 in human hepatic microsomes is mediated by multiple cytochrome p450 (p450) enzymes, in particular CYP2D6. OSU 6162 59-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-138 12433806-3 2002 Kinetics evidence is presented that the N-depropylation of (-)-OSU6162 in human hepatic microsomes is mediated by multiple cytochrome p450 (p450) enzymes, in particular CYP2D6. OSU 6162 59-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-144 12433806-10 2002 Thus, it is concluded that (-)-OSU6162 is metabolized by several p450 enzymes and that CYP2D6 accounts for the majority of the observed p450 N-depropylase activity in vitro. OSU 6162 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 12433801-4 2002 In human microsomes, inhibition of total metabolism by the CYP3A-selective inhibitors ketoconazole, troleandomycin, and 6",7"-dihydroxybergamottin, each at 10 micro M concentration, was 83 to 95%, whereas inhibition with inhibitors selective for other p450 isozymes was minimal. Ketoconazole 86-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 252-256 12433806-10 2002 Thus, it is concluded that (-)-OSU6162 is metabolized by several p450 enzymes and that CYP2D6 accounts for the majority of the observed p450 N-depropylase activity in vitro. OSU 6162 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 12433806-5 2002 incubations of (-)-OSU6162 (5 micro M) with hepatic microsomes from a panel of human donors showed that (-)-OSU6162 N-depropylase activity correlated well with CYP2D6-catalyzed dextromethorphan O-demethylase activity but not with other p450 enzyme-specific activities; 2). OSU 6162 15-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 236-240 12433801-4 2002 In human microsomes, inhibition of total metabolism by the CYP3A-selective inhibitors ketoconazole, troleandomycin, and 6",7"-dihydroxybergamottin, each at 10 micro M concentration, was 83 to 95%, whereas inhibition with inhibitors selective for other p450 isozymes was minimal. Troleandomycin 100-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 252-256 12433810-2 2002 Treatment of 2- to 3-day-old human hepatocyte cultures with 3 x 10(-5) M lovastatin, simvastatin, fluvastatin, or atorvastatin for 24 h increased the amounts of CYP2B6 and CYP3A mRNA by an average of 3.8- to 9.2-fold and 24- to 36-fold, respectively. Lovastatin 73-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 12433801-4 2002 In human microsomes, inhibition of total metabolism by the CYP3A-selective inhibitors ketoconazole, troleandomycin, and 6",7"-dihydroxybergamottin, each at 10 micro M concentration, was 83 to 95%, whereas inhibition with inhibitors selective for other p450 isozymes was minimal. 6',7'-dihydroxybergamottin 120-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 252-256 12433810-2 2002 Treatment of 2- to 3-day-old human hepatocyte cultures with 3 x 10(-5) M lovastatin, simvastatin, fluvastatin, or atorvastatin for 24 h increased the amounts of CYP2B6 and CYP3A mRNA by an average of 3.8- to 9.2-fold and 24- to 36-fold, respectively. Simvastatin 85-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 12433810-2 2002 Treatment of 2- to 3-day-old human hepatocyte cultures with 3 x 10(-5) M lovastatin, simvastatin, fluvastatin, or atorvastatin for 24 h increased the amounts of CYP2B6 and CYP3A mRNA by an average of 3.8- to 9.2-fold and 24- to 36-fold, respectively. Fluvastatin 98-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 12433810-2 2002 Treatment of 2- to 3-day-old human hepatocyte cultures with 3 x 10(-5) M lovastatin, simvastatin, fluvastatin, or atorvastatin for 24 h increased the amounts of CYP2B6 and CYP3A mRNA by an average of 3.8- to 9.2-fold and 24- to 36-fold, respectively. Atorvastatin 114-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 161-167 12433826-0 2002 Effects of olopatadine, a new antiallergic agent, on human liver microsomal cytochrome P450 activities. Olopatadine Hydrochloride 11-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 12433826-2 2002 Some compounds containing a similar alkylamino group form a cytochrome p450 (p450) -iron (II)-nitrosoalkane metabolite complex [metabolic intermediate complex (MIC)], thereby causing quasi-irreversible inhibition of the p450. iron (ii)-nitrosoalkane 84-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 12433826-2 2002 Some compounds containing a similar alkylamino group form a cytochrome p450 (p450) -iron (II)-nitrosoalkane metabolite complex [metabolic intermediate complex (MIC)], thereby causing quasi-irreversible inhibition of the p450. iron (ii)-nitrosoalkane 84-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-81 12433801-12 2002 Also, it is unlikely that EP would substantially inhibit the metabolism of other drugs that are metabolized by CYP3A4 or other p450 isoforms. Eplerenone 26-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 12433826-2 2002 Some compounds containing a similar alkylamino group form a cytochrome p450 (p450) -iron (II)-nitrosoalkane metabolite complex [metabolic intermediate complex (MIC)], thereby causing quasi-irreversible inhibition of the p450. iron (ii)-nitrosoalkane 84-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-81 12433826-4 2002 We identified the enzymes catalyzing olopatadine metabolism and investigated the effect of olopatadine on human p450 activities. Olopatadine Hydrochloride 91-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 12433826-7 2002 Incubation of olopatadine with cDNA-expressed human p450 isozymes confirmed that M1 formation was almost exclusively catalyzed by CYP3A4. Olopatadine Hydrochloride 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 12433826-10 2002 Olopatadine did not inhibit p450 activities when it was simultaneously incubated with substrates for different p450 isozymes. Olopatadine Hydrochloride 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-115 12433810-4 2002 Although treatment with the active (+), but not the inactive (-), enantiomer of atorvastatin increased the amount of HMG-CoA reductase mRNA, treatment with each enantiomer significantly induced both CYP2B6 and CYP3A mRNA levels. Atorvastatin 80-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-205 12483228-5 2002 The modeled conformation of the loop was validated by unconstrained MD simulations of the complete enzymes (CYP2C5 and CYP2B6) in water for 70 and 120 ps, respectively. Water 130-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 12451279-0 2002 Role of pharmacologic enhancement of p-450 in cyclosporine overdose. Cyclosporine 46-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-42 12386121-5 2002 Rates of carbamazepine 2- and 3-hydroxylation correlated strongly with CYP2B6 activity (r >or= 0.757) in a panel of HLMs (n = 8). Carbamazepine 9-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 12386121-8 2002 The chemical inhibitors ketoconazole (CYP3A) and 7-EFC (CYP2B6) inhibited both 2- and 3-hydroxycarbamazepine formation whereas 4-methylpyrazole (CYP2E1) markedly decreased 2-hydroxycarbamazepine formation. 2- and 3-hydroxycarbamazepine 79-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 12386121-8 2002 The chemical inhibitors ketoconazole (CYP3A) and 7-EFC (CYP2B6) inhibited both 2- and 3-hydroxycarbamazepine formation whereas 4-methylpyrazole (CYP2E1) markedly decreased 2-hydroxycarbamazepine formation. 2-hydroxycarbamazepine 172-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 12386121-10 2002 These results suggest that CYP2B6 and CYP3A4 are largely responsible for the formation of 3-hyrdoxycarbamazepine, whereas multiple P450s (CYP1A2, 2A6, 2B6, 2E1, and 3A4) contributed to 2-hydroxycarbamazepine formation. 3-hyrdoxycarbamazepine 90-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 12386121-10 2002 These results suggest that CYP2B6 and CYP3A4 are largely responsible for the formation of 3-hyrdoxycarbamazepine, whereas multiple P450s (CYP1A2, 2A6, 2B6, 2E1, and 3A4) contributed to 2-hydroxycarbamazepine formation. 2-hydroxycarbamazepine 185-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 12483228-1 2002 Human cytochrome P450 (CYP) 2B6 activates the anticancer prodrug cyclophosphamide (CPA) by 4-hydroxylation. Cyclophosphamide 65-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-31 12483228-1 2002 Human cytochrome P450 (CYP) 2B6 activates the anticancer prodrug cyclophosphamide (CPA) by 4-hydroxylation. Cyclophosphamide 83-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-31 12463733-1 2002 The clinical aspect of porphyria has been investigated, and it is well known that porphyrinogens such as estrogens and alcohol or other inducers of P450 isoenzymes exacerbate the porphyric state. Alcohols 119-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 12391268-8 2002 We conclude that the availability of heme is a limiting factor of P450 function in extrahepatic tissue. Heme 37-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 12439223-2 2002 The CYP2B6 gene has been implicated in bupropion kinetics and nicotine metabolism, and is a plausible candidate for pharmacogenetic studies of treatment response. Bupropion 39-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 12439223-2 2002 The CYP2B6 gene has been implicated in bupropion kinetics and nicotine metabolism, and is a plausible candidate for pharmacogenetic studies of treatment response. Nicotine 62-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 12434292-7 2002 The increase of mRNA amount in P450(scc) P450(c17) and 3betaHSD and the elevation of cortisol level were inhibited with a pretreatment of PD098059, a specific extracellular signal-regulated kinase inhibitor. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 138-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-49 12392414-0 2002 CYP119 plus a Sulfolobus tokodaii strain 7 ferredoxin and 2-oxoacid:ferredoxin oxidoreductase constitute a high-temperature cytochrome P450 catalytic system. 2-oxoacid 58-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-139 12392414-1 2002 The cytochrome P450 superfamily of enzymes catalyzes a broad range of oxidative processes involved in the metabolism of fatty acids, biosynthesis of sterols, and elimination of drugs and xenobiotics. Fatty Acids 120-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 12392414-1 2002 The cytochrome P450 superfamily of enzymes catalyzes a broad range of oxidative processes involved in the metabolism of fatty acids, biosynthesis of sterols, and elimination of drugs and xenobiotics. Sterols 149-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 12385721-7 2002 The production of carbaryl methylol was primarily the result of metabolism by CYP2B6. Carbaryl 18-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 12385721-11 2002 Chlorpyrifos inhibited the generation of carbaryl methylol, catalyzed predominately by CYP2B6, more than other pathways, correlating with an earlier observation that chlorpyrifos is metabolized to its oxon primarily by CYP2B6. Chlorpyrifos 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 12385721-11 2002 Chlorpyrifos inhibited the generation of carbaryl methylol, catalyzed predominately by CYP2B6, more than other pathways, correlating with an earlier observation that chlorpyrifos is metabolized to its oxon primarily by CYP2B6. Chlorpyrifos 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 219-225 12385721-11 2002 Chlorpyrifos inhibited the generation of carbaryl methylol, catalyzed predominately by CYP2B6, more than other pathways, correlating with an earlier observation that chlorpyrifos is metabolized to its oxon primarily by CYP2B6. Carbaryl 41-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 12385721-11 2002 Chlorpyrifos inhibited the generation of carbaryl methylol, catalyzed predominately by CYP2B6, more than other pathways, correlating with an earlier observation that chlorpyrifos is metabolized to its oxon primarily by CYP2B6. Carbaryl 41-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 219-225 12385721-11 2002 Chlorpyrifos inhibited the generation of carbaryl methylol, catalyzed predominately by CYP2B6, more than other pathways, correlating with an earlier observation that chlorpyrifos is metabolized to its oxon primarily by CYP2B6. Chlorpyrifos 166-178 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 12228192-2 2002 The decrease in the activity followed time- and concentration-dependent kinetics, required oxidative metabolism, and was resistant to reduced glutathione, suggesting that diclofenac causes a mechanism-based inactivation of cytochrome p450 (p450) 3A4 (CYP3A4). Glutathione 142-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 234-238 12385721-11 2002 Chlorpyrifos inhibited the generation of carbaryl methylol, catalyzed predominately by CYP2B6, more than other pathways, correlating with an earlier observation that chlorpyrifos is metabolized to its oxon primarily by CYP2B6. Chlorpyrifos 166-178 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 219-225 12228192-2 2002 The decrease in the activity followed time- and concentration-dependent kinetics, required oxidative metabolism, and was resistant to reduced glutathione, suggesting that diclofenac causes a mechanism-based inactivation of cytochrome p450 (p450) 3A4 (CYP3A4). Diclofenac 171-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 234-238 12228192-2 2002 The decrease in the activity followed time- and concentration-dependent kinetics, required oxidative metabolism, and was resistant to reduced glutathione, suggesting that diclofenac causes a mechanism-based inactivation of cytochrome p450 (p450) 3A4 (CYP3A4). Diclofenac 171-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-244 12351147-6 2002 In this work, we have re-examined the feasibility of three proposed p450 oxidation mechanisms by comparing the experimental oxidation yields and rates of 1-naphthylamine (1-NA), 2-naphthylamine (2-NA) and 2-aminofluorene (2-AF). 1-Naphthylamine 171-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 12530471-11 2002 When using chemical inhibition, however, an average inhibition percentage of 83 were obtained with orphenadrine, a human CYP2B inhibitor. Orphenadrine 99-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-126 12351147-0 2002 Metabolic oxidation of carcinogenic arylamines by p450 monooxygenases: theoretical support for the one-electron transfer mechanism. aniline 36-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 12351147-1 2002 N-oxidation by cytochrome p450 enzymes is an initial step in the metabolic activation of aromatic amine compounds. Nitrogen 0-1 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 12351147-1 2002 N-oxidation by cytochrome p450 enzymes is an initial step in the metabolic activation of aromatic amine compounds. aromatic amine 89-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 12351147-5 2002 This approach can now be improved by incorporating contemporary, ab initio quantum chemical methods to accurately determine the chemical properties of p450 aromatic amine substrates. aromatic amine 156-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-155 12351147-6 2002 In this work, we have re-examined the feasibility of three proposed p450 oxidation mechanisms by comparing the experimental oxidation yields and rates of 1-naphthylamine (1-NA), 2-naphthylamine (2-NA) and 2-aminofluorene (2-AF). 2-Naphthylamine 178-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 12351147-6 2002 In this work, we have re-examined the feasibility of three proposed p450 oxidation mechanisms by comparing the experimental oxidation yields and rates of 1-naphthylamine (1-NA), 2-naphthylamine (2-NA) and 2-aminofluorene (2-AF). 1-Naphthylamine 154-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 12351147-6 2002 In this work, we have re-examined the feasibility of three proposed p450 oxidation mechanisms by comparing the experimental oxidation yields and rates of 1-naphthylamine (1-NA), 2-naphthylamine (2-NA) and 2-aminofluorene (2-AF). 2-Naphthylamine 195-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 12351147-6 2002 In this work, we have re-examined the feasibility of three proposed p450 oxidation mechanisms by comparing the experimental oxidation yields and rates of 1-naphthylamine (1-NA), 2-naphthylamine (2-NA) and 2-aminofluorene (2-AF). 2-aminofluorene 205-220 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 12351147-9 2002 We therefore conclude, in contrast to earlier studies, that the one-electron mechanism is more likely to be the pathway for p450-catalyzed aromatic amine oxidation. aromatic amine 139-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 12167562-6 2002 A 48-h exposure of cultured human hepatocytes to bergamottin resulted in increased levels of immunoreactive CYP3A4, CYP1A1, and CYP1A2 proteins, and CYP3A4, CYP1A1, CYP1A2, CYP2B6, and UDP-glucuronosyl transferase mRNAs. bergamottin 49-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 12207498-6 2002 The kinetic parameters K(m)(app) and V(max) of the CYP1A1-dependent 8-hydroxylation activity of 2,3,7-trichloro-DD (2,3,7-triCDD) were estimated to be 0.30 microM and 51 (mol/min/mol of P450), respectively, suggesting that 2,3,7-triCDD was a good substrate for CYP1A1. 2,3,7-trichloro-dd 96-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 186-190 12207498-6 2002 The kinetic parameters K(m)(app) and V(max) of the CYP1A1-dependent 8-hydroxylation activity of 2,3,7-trichloro-DD (2,3,7-triCDD) were estimated to be 0.30 microM and 51 (mol/min/mol of P450), respectively, suggesting that 2,3,7-triCDD was a good substrate for CYP1A1. 2,3,7-tricdd 116-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 186-190 12207498-6 2002 The kinetic parameters K(m)(app) and V(max) of the CYP1A1-dependent 8-hydroxylation activity of 2,3,7-trichloro-DD (2,3,7-triCDD) were estimated to be 0.30 microM and 51 (mol/min/mol of P450), respectively, suggesting that 2,3,7-triCDD was a good substrate for CYP1A1. 2,3,7-tricdd 223-235 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 186-190 12209705-2 2002 rRp450 is a novel replication-conditional HSV-1 mutant that expresses both the endogenous herpes viral thymidine kinase gene and the rat p450 CYP2B1 transgene; p450 bioactivates such cancer prodrugs as cyclophosphamide. Cyclophosphamide 202-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 2-6 12209705-16 2002 Activation of cyclophosphamide by the p450 transgene augmented the anti-neoplastic effects of rRp450 without simultaneously decreasing viral replication. Cyclophosphamide 14-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 12167570-8 2002 Thus, CYP2B6 demethylated methoxychlor to mono-OH-M and ortho-hydroxylated the mono-OH-M forming catechol-M; however, 2B6 did not appreciably demethylate mono-OH-M or ortho-hydroxylate bis-OH-M, suggesting a narrow substrate specificity. Methoxychlor 26-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 12124303-8 2002 These two forms also catalyzed 4"-hydroxylation (13.0 +/- 1.9 and 1.4 +/- 0.1 nmol/120 min/0.2 nmol P450, respectively, for CYP2B6 and CYP2D6 at 250 microM tamoxifen; 0.51 +/- 0.08 pmol/40 min/0.2 nmol P450 for CYP2B6 at 18 microM tamoxifen). Tamoxifen 156-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-130 12167570-8 2002 Thus, CYP2B6 demethylated methoxychlor to mono-OH-M and ortho-hydroxylated the mono-OH-M forming catechol-M; however, 2B6 did not appreciably demethylate mono-OH-M or ortho-hydroxylate bis-OH-M, suggesting a narrow substrate specificity. 1,1,1-trichloro-2-(4-hydroxyphenyl)-2-(4-methoxyphenyl)ethane 42-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 12167570-8 2002 Thus, CYP2B6 demethylated methoxychlor to mono-OH-M and ortho-hydroxylated the mono-OH-M forming catechol-M; however, 2B6 did not appreciably demethylate mono-OH-M or ortho-hydroxylate bis-OH-M, suggesting a narrow substrate specificity. 1,1,1-trichloro-2-(4-hydroxyphenyl)-2-(4-methoxyphenyl)ethane 79-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 12167570-8 2002 Thus, CYP2B6 demethylated methoxychlor to mono-OH-M and ortho-hydroxylated the mono-OH-M forming catechol-M; however, 2B6 did not appreciably demethylate mono-OH-M or ortho-hydroxylate bis-OH-M, suggesting a narrow substrate specificity. catechol 97-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 12167570-8 2002 Thus, CYP2B6 demethylated methoxychlor to mono-OH-M and ortho-hydroxylated the mono-OH-M forming catechol-M; however, 2B6 did not appreciably demethylate mono-OH-M or ortho-hydroxylate bis-OH-M, suggesting a narrow substrate specificity. demethylate 13-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 12167570-8 2002 Thus, CYP2B6 demethylated methoxychlor to mono-OH-M and ortho-hydroxylated the mono-OH-M forming catechol-M; however, 2B6 did not appreciably demethylate mono-OH-M or ortho-hydroxylate bis-OH-M, suggesting a narrow substrate specificity. 1,1,1-trichloro-2-(4-hydroxyphenyl)-2-(4-methoxyphenyl)ethane 79-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 12167570-8 2002 Thus, CYP2B6 demethylated methoxychlor to mono-OH-M and ortho-hydroxylated the mono-OH-M forming catechol-M; however, 2B6 did not appreciably demethylate mono-OH-M or ortho-hydroxylate bis-OH-M, suggesting a narrow substrate specificity. bis-oh-m 185-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 12207635-5 2002 RESULTS: Z-4-hydroxy-tamoxifen was formed by supersomes expressing CYP2B6, CYP2C9, CYP2C19 and CYP2D6, but not CYP3A4. z-4-hydroxy-tamoxifen 9-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 12207635-6 2002 In agreement with these data, the mean formation of Z-4-hydroxy-tamoxifen was inhibited 49% by sulphaphenazole (P=0.001), 38% by quinidine (P<0.05) and 13% by monoclonal antibody against CYP2B6 (MAB-2B6, P<0.05). z-4-hydroxy-tamoxifen 52-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 190-196 12207635-10 2002 CONCLUSIONS: CYP2B6, CYP2C9 and CYP2D6 genotypes all affected Z-4-hydroxy-tamoxifen formation and can predict individual ability to catalyse this reaction. z-4-hydroxy-tamoxifen 62-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 12160905-6 2002 Activation of benzhydrol and p-benzoylphenol by the P450/NPR system was similar to that of benzophenone. benzohydrol 14-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-60 12160905-6 2002 Activation of benzhydrol and p-benzoylphenol by the P450/NPR system was similar to that of benzophenone. 4-hydroxybenzophenone 29-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-60 12045201-4 2002 The cholesterol-sensing liver X receptor competes with CXR, pregnane X receptor, or constitutive androstane receptor for regulation of drug-responsive enhancers from chicken CYP2H1, human CYP3A4, or human CYP2B6, respectively. Cholesterol 4-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 205-211 12167460-0 2002 Effect of cyclophosphamide on gene expression of cytochromes p450 and beta-actin in the HL-60 cell line. Cyclophosphamide 10-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 12207635-0 2002 The influence of CYP2B6, CYP2C9 and CYP2D6 genotypes on the formation of the potent antioestrogen Z-4-hydroxy-tamoxifen in human liver. z-4-hydroxy-tamoxifen 98-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 12207635-2 2002 METHODS: The formation of Z-4-hydroxy-tamoxifen from 10 microm tamoxifen was studied in human liver microsomes (n=50), characterized for CYP2B6, CYP2C9, CYP2D6 and CYP3A4 expression, and CYP2B6, CYP2C9 and CYP2D6 genotype. z-4-hydroxy-tamoxifen 26-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 12207635-2 2002 METHODS: The formation of Z-4-hydroxy-tamoxifen from 10 microm tamoxifen was studied in human liver microsomes (n=50), characterized for CYP2B6, CYP2C9, CYP2D6 and CYP3A4 expression, and CYP2B6, CYP2C9 and CYP2D6 genotype. z-4-hydroxy-tamoxifen 26-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 12124303-8 2002 These two forms also catalyzed 4"-hydroxylation (13.0 +/- 1.9 and 1.4 +/- 0.1 nmol/120 min/0.2 nmol P450, respectively, for CYP2B6 and CYP2D6 at 250 microM tamoxifen; 0.51 +/- 0.08 pmol/40 min/0.2 nmol P450 for CYP2B6 at 18 microM tamoxifen). Tamoxifen 156-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 211-217 12124303-8 2002 These two forms also catalyzed 4"-hydroxylation (13.0 +/- 1.9 and 1.4 +/- 0.1 nmol/120 min/0.2 nmol P450, respectively, for CYP2B6 and CYP2D6 at 250 microM tamoxifen; 0.51 +/- 0.08 pmol/40 min/0.2 nmol P450 for CYP2B6 at 18 microM tamoxifen). Tamoxifen 231-240 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-130 12124303-8 2002 These two forms also catalyzed 4"-hydroxylation (13.0 +/- 1.9 and 1.4 +/- 0.1 nmol/120 min/0.2 nmol P450, respectively, for CYP2B6 and CYP2D6 at 250 microM tamoxifen; 0.51 +/- 0.08 pmol/40 min/0.2 nmol P450 for CYP2B6 at 18 microM tamoxifen). Tamoxifen 231-240 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 211-217 11991950-1 2002 The fully active dihydroxylated metabolite of vitamin D(3) induces the expression of CYP3A4 and, to a lesser extent, CYP2B6 and CYP2C9 genes in normal differentiated primary human hepatocytes. Vitamin D 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 12061800-3 2002 These results indicate that 2-PMADA and 3-PMDIA belong among the most potent CYP2B6-selective inhibitors discovered to date. 2-isopropenyl-2-methyladamantane 28-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 11997390-0 2002 Cytochrome P450 omega-hydroxylase pathway of tocopherol catabolism. Tocopherols 45-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 11997390-3 2002 Here we describe a pathway involving cytochrome P450-mediated omega-hydroxylation of the tocopherol phytyl side chain followed by stepwise removal of two- or three-carbon moieties, ultimately yielding the 3"-carboxychromanol metabolite that is excreted in urine. Tocopherols 89-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 11997390-3 2002 Here we describe a pathway involving cytochrome P450-mediated omega-hydroxylation of the tocopherol phytyl side chain followed by stepwise removal of two- or three-carbon moieties, ultimately yielding the 3"-carboxychromanol metabolite that is excreted in urine. Carbon 164-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 11997390-3 2002 Here we describe a pathway involving cytochrome P450-mediated omega-hydroxylation of the tocopherol phytyl side chain followed by stepwise removal of two- or three-carbon moieties, ultimately yielding the 3"-carboxychromanol metabolite that is excreted in urine. 3"-carboxychromanol 205-224 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 11997390-6 2002 Functional analysis of several recombinant human liver P450 enzymes revealed that tocopherol-omega-hydroxylase activity was associated only with CYP4F2, which also catalyzes omega-hydroxylation of leukotriene B(4) and arachidonic acid. Leukotriene B4 197-210 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 11997390-6 2002 Functional analysis of several recombinant human liver P450 enzymes revealed that tocopherol-omega-hydroxylase activity was associated only with CYP4F2, which also catalyzes omega-hydroxylation of leukotriene B(4) and arachidonic acid. Arachidonic Acid 218-234 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 12061800-3 2002 These results indicate that 2-PMADA and 3-PMDIA belong among the most potent CYP2B6-selective inhibitors discovered to date. 3-isopropenyl-3-methyldiamantane 40-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 12065440-1 2002 The purpose of this study was to characterize the concentration-response effects of cyclophosphamide (CPA) with and without dexamethasone (DEX; 10 microM) on the expression of CYP3A4 and CYP2B6 in cultured human hepatocytes at concentrations representative of standard- and high-dose CPA therapy (25 to 750 microM). Cyclophosphamide 84-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 12065445-0 2002 Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Ketamine 74-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 12065437-0 2002 Identification of human hepatic cytochrome P450 sources of N-alkylprotoporphyrin IX after interaction with porphyrinogenic xenobiotics, implications for detection of xenobiotic-induced porphyria in humans. n-alkylprotoporphyrin ix 59-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 12065445-7 2002 N-demethylation of ketamine was correlated with nifedipine oxidase activity (CYP3A4-specific marker reaction), and it was also correlated with S-mephenytoin N-demethylase activity (CYP2B6-specific marker reaction). Ketamine 19-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-187 12065440-1 2002 The purpose of this study was to characterize the concentration-response effects of cyclophosphamide (CPA) with and without dexamethasone (DEX; 10 microM) on the expression of CYP3A4 and CYP2B6 in cultured human hepatocytes at concentrations representative of standard- and high-dose CPA therapy (25 to 750 microM). Cyclophosphamide 102-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 12065445-9 2002 In human lymphoblast-expressed P450, the activities of CYP2B6 were higher than those of CYP3A4 and CYP2C9 at three concentrations of ketamine, 0.005, 0.05, and 0.5 mM. Ketamine 133-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 12065437-1 2002 Porphyrinogenicity of certain xenobiotics depends upon mechanism-based inactivation of specific cytochrome P450 (P450) enzymes, followed by formation of N-alkylprotoporphyrin IX (N-alkylPP). n-alkylprotoporphyrin ix 153-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-111 12065437-1 2002 Porphyrinogenicity of certain xenobiotics depends upon mechanism-based inactivation of specific cytochrome P450 (P450) enzymes, followed by formation of N-alkylprotoporphyrin IX (N-alkylPP). n-alkylprotoporphyrin ix 153-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-117 12065440-1 2002 The purpose of this study was to characterize the concentration-response effects of cyclophosphamide (CPA) with and without dexamethasone (DEX; 10 microM) on the expression of CYP3A4 and CYP2B6 in cultured human hepatocytes at concentrations representative of standard- and high-dose CPA therapy (25 to 750 microM). Dexamethasone 124-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 12065437-1 2002 Porphyrinogenicity of certain xenobiotics depends upon mechanism-based inactivation of specific cytochrome P450 (P450) enzymes, followed by formation of N-alkylprotoporphyrin IX (N-alkylPP). n-alkylpp 179-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-111 12065445-11 2002 The present study demonstrates that CYP3A4 is the principal enzyme responsible for ketamine N-demethylation in human liver microsomes and that CYP2B6 and CYP2C9 have a minor contribution to ketamine N-demethylation at therapeutic concentrations of the drug. ketamine n 190-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 12065437-1 2002 Porphyrinogenicity of certain xenobiotics depends upon mechanism-based inactivation of specific cytochrome P450 (P450) enzymes, followed by formation of N-alkylprotoporphyrin IX (N-alkylPP). n-alkylpp 179-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-117 12065440-1 2002 The purpose of this study was to characterize the concentration-response effects of cyclophosphamide (CPA) with and without dexamethasone (DEX; 10 microM) on the expression of CYP3A4 and CYP2B6 in cultured human hepatocytes at concentrations representative of standard- and high-dose CPA therapy (25 to 750 microM). Dexamethasone 139-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 12065440-1 2002 The purpose of this study was to characterize the concentration-response effects of cyclophosphamide (CPA) with and without dexamethasone (DEX; 10 microM) on the expression of CYP3A4 and CYP2B6 in cultured human hepatocytes at concentrations representative of standard- and high-dose CPA therapy (25 to 750 microM). Cyclophosphamide 284-287 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 12065440-2 2002 CPA produced concentration-dependent increases in CYP3A4 and CYP2B6 activity and immunoreactive protein that peaked at 250 and 125 microM, respectively, and declined thereafter. Cyclophosphamide 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 12065440-3 2002 The inductive effect of CPA alone and in combination with DEX was greater in magnitude for CYP2B6 compared with CYP3A4. Cyclophosphamide 24-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 12065440-3 2002 The inductive effect of CPA alone and in combination with DEX was greater in magnitude for CYP2B6 compared with CYP3A4. Dexamethasone 58-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 12084621-3 2002 Isozymes selective for imidazolidine oxidation in order of decreasing overall activity are CYP3A4>CYP2C19 or CYP2A6>CYP2C9, while those selective for nitroimine reduction are CYP1A2, CYP2B6, CYP2D6 and CYP2E1. Imidazolidines 23-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-195 12162853-0 2002 Involvement of cytochrome P450 in the metabolism of rebamipide by the human liver. rebamipide 52-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 12162853-2 2002 The metabolism of rebamipide, a gastroprotective agent, was investigated using human liver microsomes and cDNA-expressed human cytochrome P450 systems. rebamipide 18-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-142 12162853-14 2002 It is therefore considered that drug interactions with cytochrome P450 enzymes are not involved in either the metabolism of rebamipide or the metabolism of other drugs concomitantly administered with rebamipide. rebamipide 124-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 12162853-14 2002 It is therefore considered that drug interactions with cytochrome P450 enzymes are not involved in either the metabolism of rebamipide or the metabolism of other drugs concomitantly administered with rebamipide. rebamipide 200-210 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 12059209-0 2002 Evaluation of norcarane as a probe for radicals in cytochome p450- and soluble methane monooxygenase-catalyzed hydroxylation reactions. norcarane 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 12059209-1 2002 Norcarane was employed as a mechanistic probe in oxidations catalyzed by hepatic cytochome P450 enzymes and by the soluble methane monooxygenase (sMMO) enzyme from Methylococcuscapsulatus (Bath). norcarane 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 12059209-9 2002 We conclude that generalizations regarding the intermediacy of radicals in P450 and sMMO enzyme-catalyzed hydroxylations based on the norcarane results should be considered hypothetical until the origin of the unanticipated results can be determined. norcarane 134-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 12084621-3 2002 Isozymes selective for imidazolidine oxidation in order of decreasing overall activity are CYP3A4>CYP2C19 or CYP2A6>CYP2C9, while those selective for nitroimine reduction are CYP1A2, CYP2B6, CYP2D6 and CYP2E1. nitroimine 156-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-195 12050123-12 2002 A varied prevalence of hypergonadotropic hypogonadism in patients with AD and value of steroid-producing cells autoantibodies reactive with steroid 17alpha-hydroxylase or P450 side-chain cleavage enzyme as markers of this disease has been discussed. Steroids 87-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-175 12107550-7 2002 Cloned, expressed CYP2B6 and CYP3A4 microsomes catalyzed TEPA formation, whereas CYP2A6 and CYP2E1 did not. Triethylenephosphoramide 57-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 12107550-8 2002 Incubation of thiotepa with anti-CYP2B6 antibody and cloned, expressed CYP2B6 microsomes resulted in reductions in the formation of TEPA, but no change in TEPA formation occurred in human liver microsomes. Thiotepa 14-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 12107550-8 2002 Incubation of thiotepa with anti-CYP2B6 antibody and cloned, expressed CYP2B6 microsomes resulted in reductions in the formation of TEPA, but no change in TEPA formation occurred in human liver microsomes. Thiotepa 14-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 12107550-8 2002 Incubation of thiotepa with anti-CYP2B6 antibody and cloned, expressed CYP2B6 microsomes resulted in reductions in the formation of TEPA, but no change in TEPA formation occurred in human liver microsomes. Triethylenephosphoramide 132-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 12107550-8 2002 Incubation of thiotepa with anti-CYP2B6 antibody and cloned, expressed CYP2B6 microsomes resulted in reductions in the formation of TEPA, but no change in TEPA formation occurred in human liver microsomes. Triethylenephosphoramide 132-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 12107550-9 2002 CONCLUSIONS: Thiotepa is metabolized in human liver microsomes by CYP3A4 (major) and CYP2B6 (minor). Thiotepa 13-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 12160484-14 2002 Lower rates of V11294 metabolism to some V11294 metabolites were also observed with cDNA-expressed CYP2C9, CYP2C19 and CYP2D6, whereas only very low or undetectable rates of V11294 metabolism were observed with cDNA-expressed CYP1A2, CYP2A6, CYP2B6, CYP2C8 and CYP2E1. 3-(3-cyclopentyloxy-4-methoxybenzyl)-6-ethylamino-8-isopropyl-3H-purine 15-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-248 12023523-0 2002 Effect of tamoxifen on the enzymatic activity of human cytochrome CYP2B6. Tamoxifen 10-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-72 12023523-4 2002 The effect of tamoxifen on the enzymatic activity of bacterially expressed human cytochrome CYP2B6 in a reconstituted system has been investigated. Tamoxifen 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 12023523-5 2002 The 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation activity of purified CYP2B6 was inactivated by tamoxifen in a time- and concentration-dependent manner. 7-ethoxy-4-trifluoromethylcoumarin 4-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 12023523-5 2002 The 7-ethoxy-4-(trifluoromethyl)coumarin O-deethylation activity of purified CYP2B6 was inactivated by tamoxifen in a time- and concentration-dependent manner. Tamoxifen 103-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 12023523-9 2002 The activity of CYP2B6 was not recovered after removal of free tamoxifen using spin column gel filtration. Tamoxifen 63-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 12023523-11 2002 A reconstituted system containing purified CYP2B6, NADPH-reductase, and NADPH-generating system was found to catalyze tamoxifen metabolism to 4-OH-tamoxifen, 4"-OH-tamoxifen, and N-desmethyl-tamoxifen as analyzed by high-performance liquid chromatography analysis. Tamoxifen 118-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 12023523-11 2002 A reconstituted system containing purified CYP2B6, NADPH-reductase, and NADPH-generating system was found to catalyze tamoxifen metabolism to 4-OH-tamoxifen, 4"-OH-tamoxifen, and N-desmethyl-tamoxifen as analyzed by high-performance liquid chromatography analysis. 4-oh-tamoxifen 142-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 12023523-11 2002 A reconstituted system containing purified CYP2B6, NADPH-reductase, and NADPH-generating system was found to catalyze tamoxifen metabolism to 4-OH-tamoxifen, 4"-OH-tamoxifen, and N-desmethyl-tamoxifen as analyzed by high-performance liquid chromatography analysis. 4"-oh-tamoxifen 158-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 12023523-11 2002 A reconstituted system containing purified CYP2B6, NADPH-reductase, and NADPH-generating system was found to catalyze tamoxifen metabolism to 4-OH-tamoxifen, 4"-OH-tamoxifen, and N-desmethyl-tamoxifen as analyzed by high-performance liquid chromatography analysis. N-desmethyltamoxifen 179-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 12019191-6 2002 P450 isoforms mediating sildenafil biotransformation differ substantially between humans and the male rat, thereby limiting the applicability of this species as a model for sildenafil metabolism and drug interactions in humans. Sildenafil Citrate 24-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 11933021-2 2002 Nonsteroidal aromatase inhibitors (NSAIs) are competitive inhibitors of aromatase, which bind to the enzyme active site by coordinating the iron atom present in the heme group of the P450 protein. Iron 140-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 12022835-3 2002 Products indicative of a radical intermediate with a lifetime ranging from 16 to 52 ps were detected during the oxidation of norcarane by P450(cam) (CYP101), P450(BM3) (CYP102), CYP2B1, and CYP2E1. norcarane 125-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-167 11950794-6 2002 Of 11 recombinant human P450 enzymes (expressed in Trichoplusia ni cells) tested, CYP2C8, 2C9, 2C18, 2C19, and CYP3A4 catalyzed oxidations of (+)- and (-)-limonenes to respective carveols and perillyl alcohol. (+)- and (-)-limonenes 142-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 11950794-6 2002 Of 11 recombinant human P450 enzymes (expressed in Trichoplusia ni cells) tested, CYP2C8, 2C9, 2C18, 2C19, and CYP3A4 catalyzed oxidations of (+)- and (-)-limonenes to respective carveols and perillyl alcohol. carveol 179-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 11950794-6 2002 Of 11 recombinant human P450 enzymes (expressed in Trichoplusia ni cells) tested, CYP2C8, 2C9, 2C18, 2C19, and CYP3A4 catalyzed oxidations of (+)- and (-)-limonenes to respective carveols and perillyl alcohol. perillyl alcohol 192-208 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 11950794-7 2002 Interestingly, human CYP2B6 did not catalyze limonene oxidations, whereas rat CYP2B1 had high activities in catalyzing limonene oxidations. Limonene 119-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 12088278-1 2002 The demonstration of a role for microsomal P450 in the metabolism of endogenous pools of arachidonic acid established this enzyme system as a member of the arachidonic acid cascade and characterized a new an important metabolic function for this enzyme system. Arachidonic Acid 89-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 12088278-1 2002 The demonstration of a role for microsomal P450 in the metabolism of endogenous pools of arachidonic acid established this enzyme system as a member of the arachidonic acid cascade and characterized a new an important metabolic function for this enzyme system. Arachidonic Acid 156-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 12088278-2 2002 Studies from several laboratories documenting the powerful biological activities of the P450-derived eicosanoids have suggested important roles for the P450 arachidonic acid monooxygenase in renal and vascular physiology, and in the pathophysiology of experimental hypertension. Eicosanoids 101-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 12088278-2 2002 Studies from several laboratories documenting the powerful biological activities of the P450-derived eicosanoids have suggested important roles for the P450 arachidonic acid monooxygenase in renal and vascular physiology, and in the pathophysiology of experimental hypertension. Eicosanoids 101-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-156 11950782-0 2002 Triethylenethiophosphoramide is a specific inhibitor of cytochrome P450 2B6: implications for cyclophosphamide metabolism. Thiotepa 0-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-75 11950782-0 2002 Triethylenethiophosphoramide is a specific inhibitor of cytochrome P450 2B6: implications for cyclophosphamide metabolism. Cyclophosphamide 94-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-75 11950782-4 2002 Studies suggest that CYP2B6 plays an important role in the 4-hydroxylation of cyclophosphamide and that this reaction can be inhibited by triethylenethiophosphoramide (thioTEPA). Cyclophosphamide 78-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 11950782-4 2002 Studies suggest that CYP2B6 plays an important role in the 4-hydroxylation of cyclophosphamide and that this reaction can be inhibited by triethylenethiophosphoramide (thioTEPA). Thiotepa 138-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 11950782-4 2002 Studies suggest that CYP2B6 plays an important role in the 4-hydroxylation of cyclophosphamide and that this reaction can be inhibited by triethylenethiophosphoramide (thioTEPA). Thiotepa 168-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 11950782-5 2002 We therefore wished to test the hypothesis that thioTEPA is an inhibitor of CYP2B6. Thiotepa 48-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 11950782-6 2002 Using human liver microsomes (HLMs) and recombinant P450 enzymes, we demonstrated that thioTEPA is a potent and specific inhibitor of CYP2B6. Thiotepa 87-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-140 11950782-7 2002 Enzyme activity was reduced 78.1 +/- 0.2% by 50 microM thioTEPA when CYP2B6 activity was measured by following the metabolism of 200 microM S-mephenytoin to nirvanol. Thiotepa 55-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 11950782-7 2002 Enzyme activity was reduced 78.1 +/- 0.2% by 50 microM thioTEPA when CYP2B6 activity was measured by following the metabolism of 200 microM S-mephenytoin to nirvanol. Mephenytoin 140-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 11950782-7 2002 Enzyme activity was reduced 78.1 +/- 0.2% by 50 microM thioTEPA when CYP2B6 activity was measured by following the metabolism of 200 microM S-mephenytoin to nirvanol. ethylphenylhydantoin 157-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 11950782-9 2002 thioTEPA seems to be a potent noncompetitive inhibitor of CYP2B6, with K(i) values of 4.8 +/- 0.3 and 6.2 +/- 0.7 microM for HLMs and recombinant CYP2B6, respectively, values that are within the plasma concentration range of thioTEPA at therapeutic doses (1.1-18.6 microM). Thiotepa 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 11950782-9 2002 thioTEPA seems to be a potent noncompetitive inhibitor of CYP2B6, with K(i) values of 4.8 +/- 0.3 and 6.2 +/- 0.7 microM for HLMs and recombinant CYP2B6, respectively, values that are within the plasma concentration range of thioTEPA at therapeutic doses (1.1-18.6 microM). Thiotepa 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 146-152 11950782-9 2002 thioTEPA seems to be a potent noncompetitive inhibitor of CYP2B6, with K(i) values of 4.8 +/- 0.3 and 6.2 +/- 0.7 microM for HLMs and recombinant CYP2B6, respectively, values that are within the plasma concentration range of thioTEPA at therapeutic doses (1.1-18.6 microM). Thiotepa 225-233 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 11950782-10 2002 We conclude that thioTEPA is a potent and specific inhibitor of CYP2B6 and that this is the likely mechanism by which thioTEPA inhibits the activation of cyclophosphamide. Thiotepa 17-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 11950782-10 2002 We conclude that thioTEPA is a potent and specific inhibitor of CYP2B6 and that this is the likely mechanism by which thioTEPA inhibits the activation of cyclophosphamide. Thiotepa 118-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 11950782-10 2002 We conclude that thioTEPA is a potent and specific inhibitor of CYP2B6 and that this is the likely mechanism by which thioTEPA inhibits the activation of cyclophosphamide. Cyclophosphamide 154-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-70 11933021-2 2002 Nonsteroidal aromatase inhibitors (NSAIs) are competitive inhibitors of aromatase, which bind to the enzyme active site by coordinating the iron atom present in the heme group of the P450 protein. Heme 165-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 11854147-9 2002 Preincubation of human CYP2B6 with chlorpyrifos completely inhibited the metabolism of DEET. Chlorpyrifos 35-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 12040753-6 2002 The levels of CYP1A1, CYP2B6, CYP2C8, CYP3A4, CYP3A5, ADH3, and ABCG1 mRNA in human hepatocytes increased after exposure to rifampicin. Rifampin 124-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 12020135-0 2002 Use of kinetic isotope effects to delineate the role of phenylalanine 87 in P450(BM-3). Phenylalanine 56-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-85 12020135-5 2002 Based on crystal structures and the fact that the P450(BM-3) F87A mutant produces a large isotope in contrast to the native enzyme, we propose that phenylalanine 87 is responsible for hindering substrate access to the active oxygen species for nonnative substrates. Phenylalanine 148-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-59 12020135-5 2002 Based on crystal structures and the fact that the P450(BM-3) F87A mutant produces a large isotope in contrast to the native enzyme, we propose that phenylalanine 87 is responsible for hindering substrate access to the active oxygen species for nonnative substrates. Oxygen 225-231 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-59 11901092-1 2002 The inhibition and mechanism-based inactivation potencies of irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin; CPT-11) and its active metabolite (7-ethyl-10-hydroxycamptothecin; SN-38) for human cytochrome P450 (P450) enzymes were investigated to evaluate the potential for drug interactions involving CPT-11 using microsomes from insect cells expressing specific human P450 isoforms. Irinotecan 73-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 224-245 11854139-2 2002 In the present study, N-3-benzyl derivatives of nirvanol and phenobarbital were synthesized, their respective (+)- and (-)-enantiomers resolved chromatographically, and inhibitor potencies determined for these compounds toward CYP2C19 and other human liver cytochromes P450 (P450s). ethylphenylhydantoin 48-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 269-273 11854139-2 2002 In the present study, N-3-benzyl derivatives of nirvanol and phenobarbital were synthesized, their respective (+)- and (-)-enantiomers resolved chromatographically, and inhibitor potencies determined for these compounds toward CYP2C19 and other human liver cytochromes P450 (P450s). Phenobarbital 61-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 269-273 11854155-2 2002 Using human liver microsomes (HLMs) and recombinant human cytochromes P450 (P450), we identified the major route of metoclopramide oxidation and the P450 isoforms involved. Metoclopramide 116-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 11854155-2 2002 Using human liver microsomes (HLMs) and recombinant human cytochromes P450 (P450), we identified the major route of metoclopramide oxidation and the P450 isoforms involved. Metoclopramide 116-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 11854155-2 2002 Using human liver microsomes (HLMs) and recombinant human cytochromes P450 (P450), we identified the major route of metoclopramide oxidation and the P450 isoforms involved. Metoclopramide 116-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 11854155-3 2002 We also documented the ability of metoclopramide to inhibit the P450 system, using isoform-specific substrate reaction probes of CYP1A2, 2C19, 2C9, 2D6, 2E1, and 3A4. Metoclopramide 34-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 11854155-8 2002 Among the recombinant human P450 isoforms examined, monodeethylmetoclopramide was formed at the highest rate by CYP2D6 (V = 4.5 +/- 0.3 pmol/min/pmol of P450) and to a lesser extent by CYP1A2 (0.97 +/- 0.15 pmol/min/pmol of P450). monodeethylmetoclopramide 52-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-32 11854155-8 2002 Among the recombinant human P450 isoforms examined, monodeethylmetoclopramide was formed at the highest rate by CYP2D6 (V = 4.5 +/- 0.3 pmol/min/pmol of P450) and to a lesser extent by CYP1A2 (0.97 +/- 0.15 pmol/min/pmol of P450). monodeethylmetoclopramide 52-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 11854155-8 2002 Among the recombinant human P450 isoforms examined, monodeethylmetoclopramide was formed at the highest rate by CYP2D6 (V = 4.5 +/- 0.3 pmol/min/pmol of P450) and to a lesser extent by CYP1A2 (0.97 +/- 0.15 pmol/min/pmol of P450). monodeethylmetoclopramide 52-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 12373745-5 2002 Pulsed ultrafiltration can also be used in conjunction with LC-MS-MS to screen mixtures for compounds that might be activated metabolically to electrophilic quinoid and epoxide metabolites by cytochrome p450; that screening can provide early warning of compounds likely to be toxic when administered in large doses. quinoid 157-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 203-207 12373745-5 2002 Pulsed ultrafiltration can also be used in conjunction with LC-MS-MS to screen mixtures for compounds that might be activated metabolically to electrophilic quinoid and epoxide metabolites by cytochrome p450; that screening can provide early warning of compounds likely to be toxic when administered in large doses. Epoxy Compounds 169-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 203-207 11889205-3 2002 The enzyme aromatase P450 is responsible for the conversion of androgen precursor steroids to estrogens and may, therefore, have a role in regulating adipose tissue mass and its distribution. Steroids 82-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 11852064-1 2002 A simple approach to study the activity and stoichiometry of cytochrome P-450 IIB1-catalyzed metabolism of pentoxyresorufin (PRF) has been investigated. pentoxyresorufin 107-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-82 11792676-1 2002 There is very limited information on cytochrome P450 (P450)-mediated oxidative metabolism of dietary flavonoids in humans. Flavonoids 101-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 11792676-1 2002 There is very limited information on cytochrome P450 (P450)-mediated oxidative metabolism of dietary flavonoids in humans. Flavonoids 101-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 11792676-2 2002 In this study, we used human liver microsomes and recombinant P450 isoforms to examine the metabolism of two flavonols, galangin and kaempferide, and one flavone, chrysin. Flavonols 109-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 11792676-5 2002 Consistent with these observations, the human liver microsomal metabolism of galangin and kaempferide were inhibited by the P450 inhibitors furafylline and sulfaphenazole. kaempferide 90-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 11792676-5 2002 Consistent with these observations, the human liver microsomal metabolism of galangin and kaempferide were inhibited by the P450 inhibitors furafylline and sulfaphenazole. furafylline 140-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 11792676-5 2002 Consistent with these observations, the human liver microsomal metabolism of galangin and kaempferide were inhibited by the P450 inhibitors furafylline and sulfaphenazole. Sulfaphenazole 156-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 11871398-8 2002 Measurements of kinetic parameters of P450 enzymes that could metabolize both steroids, combined with the fact that CYP3A4 is known to be the most abundant P450 enzyme in the human liver, indicate that CYP3A4 will be of major importance for the in vivo human metabolism of Org 4060 and Org 30659. Steroids 78-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 11796529-8 2002 This inhibitory effect was associated with significant decreases in human CG-stimulated expression levels of the mRNAs encoding steroid acute regulatory protein, and P450 cholesterol side-chain cleavage, 3beta-hydroxy steroid dehydrogenase, and 17beta-hydroxy steroid dehydrogenase type III enzymes. cysteinylglycine 74-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-170 11871398-8 2002 Measurements of kinetic parameters of P450 enzymes that could metabolize both steroids, combined with the fact that CYP3A4 is known to be the most abundant P450 enzyme in the human liver, indicate that CYP3A4 will be of major importance for the in vivo human metabolism of Org 4060 and Org 30659. Steroids 78-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 156-160 11744616-6 2002 Benzyloxyresorufin and 7-ethoxy-4-trifluoromethylcoumarin, the two lowest K(m) substrates in the training set, were then docked in the active site of CYP2B6. benzyloxyresorufin 0-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 11744616-6 2002 Benzyloxyresorufin and 7-ethoxy-4-trifluoromethylcoumarin, the two lowest K(m) substrates in the training set, were then docked in the active site of CYP2B6. 7-ethoxy-4-trifluoromethylcoumarin 23-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-156 11744616-9 2002 The pharmacophores and the CYP2B6 model were used in conjunction to predict the K(m) values of substrates in a test set of five compounds and yielded satisfactory predictions for benzphetamine, cinnarizine, bupropion, and verapamil but not lidocaine. Benzphetamine 179-192 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 11744616-9 2002 The pharmacophores and the CYP2B6 model were used in conjunction to predict the K(m) values of substrates in a test set of five compounds and yielded satisfactory predictions for benzphetamine, cinnarizine, bupropion, and verapamil but not lidocaine. Cinnarizine 194-205 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 11744616-9 2002 The pharmacophores and the CYP2B6 model were used in conjunction to predict the K(m) values of substrates in a test set of five compounds and yielded satisfactory predictions for benzphetamine, cinnarizine, bupropion, and verapamil but not lidocaine. Bupropion 207-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 11744616-9 2002 The pharmacophores and the CYP2B6 model were used in conjunction to predict the K(m) values of substrates in a test set of five compounds and yielded satisfactory predictions for benzphetamine, cinnarizine, bupropion, and verapamil but not lidocaine. Verapamil 222-231 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 11744616-9 2002 The pharmacophores and the CYP2B6 model were used in conjunction to predict the K(m) values of substrates in a test set of five compounds and yielded satisfactory predictions for benzphetamine, cinnarizine, bupropion, and verapamil but not lidocaine. Lidocaine 240-249 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 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 2 subfamily B member 6 Homo sapiens 90-96 11876499-6 2002 In cultured hepatocytes, astaxanthin was a significant inducer of the major cytochrome P450 enzyme, CYP3A4 as well as of CYP2B6, but not of other CYPs, including those from CYP1A and CYP2C families. astaxanthine 25-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-127 12751910-4 2002 Quantitative Structure-Activity Relationships (QSARs) for substrates binding to CYP2B6 indicate a key role for hydrogen bonding, and lipophilic character, as determined by the log P parameter (where P is the octanol/water partition coefficient), is also of importance for explaining the variation in experimental binding affinity for CYP2B6 substrates. Hydrogen 111-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 12751910-4 2002 Quantitative Structure-Activity Relationships (QSARs) for substrates binding to CYP2B6 indicate a key role for hydrogen bonding, and lipophilic character, as determined by the log P parameter (where P is the octanol/water partition coefficient), is also of importance for explaining the variation in experimental binding affinity for CYP2B6 substrates. Octanols 208-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 12751910-4 2002 Quantitative Structure-Activity Relationships (QSARs) for substrates binding to CYP2B6 indicate a key role for hydrogen bonding, and lipophilic character, as determined by the log P parameter (where P is the octanol/water partition coefficient), is also of importance for explaining the variation in experimental binding affinity for CYP2B6 substrates. Water 216-221 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 15618670-3 2002 CYP1A2 and CYP3A4 also contributed to the metabolism of selegiline but their catalytic activities were much less than that of CYP2B6. Selegiline 56-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 15618670-4 2002 CYP2B6 had a higher affinity for both N-depropagylation (K(m)=21.4 microM) and N-demethylation (K(m)=25.2 microM) of selegiline than CYP3A4 and CYP1A2. Nitrogen 38-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15618670-4 2002 CYP2B6 had a higher affinity for both N-depropagylation (K(m)=21.4 microM) and N-demethylation (K(m)=25.2 microM) of selegiline than CYP3A4 and CYP1A2. Selegiline 117-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 15618670-5 2002 In immunoinhibition studies using mixed human hepatic microsomes, selegiline N-depropagylation activity was most strongly inhibited by anti-CYP2B and anti-CYP3A antibodies, while selegiline N-demethylation activity was most inhibited by anti-CYP2B antibody. selegiline n 66-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-145 15618670-5 2002 In immunoinhibition studies using mixed human hepatic microsomes, selegiline N-depropagylation activity was most strongly inhibited by anti-CYP2B and anti-CYP3A antibodies, while selegiline N-demethylation activity was most inhibited by anti-CYP2B antibody. selegiline n 66-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 242-247 15618670-7 2002 Selegiline inhibited CYP2B6-mediated (S)-mephenytoin N-demethylation activity and CYP2C19-mediated (S)-mephenytoin 4"-hydroxylation activity. Selegiline 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 15618670-7 2002 Selegiline inhibited CYP2B6-mediated (S)-mephenytoin N-demethylation activity and CYP2C19-mediated (S)-mephenytoin 4"-hydroxylation activity. Mephenytoin 37-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 15618670-9 2002 In conclusion, CYP2B6 participates in the metabolism of selegiline but the degree of its contribution varies with the level of its expression in human liver. Selegiline 56-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 12688525-0 2002 Organic nitrate tolerance is induced by degradation of some cytochrome P450 isoforms. organic nitrate 0-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 12688525-6 2002 At 48 h after infusion, NTG-induced NO generation of the vessels increased in acetone (a P450 inducer)-pretreated rats, and nitrite and nitrate levels were markedly greater than in normal rats. Nitroglycerin 24-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-93 12688525-2 2002 We studied the rat P450 (CYP)-catalyzed conversion of organic nitrate to nitric oxide (NO) by purified CYP isoforms and the relationship between P450 expression and nitrate tolerance following continuous infusion of organic nitrates in rats. Nitrates 62-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 12688525-6 2002 At 48 h after infusion, NTG-induced NO generation of the vessels increased in acetone (a P450 inducer)-pretreated rats, and nitrite and nitrate levels were markedly greater than in normal rats. Acetone 78-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-93 12688525-2 2002 We studied the rat P450 (CYP)-catalyzed conversion of organic nitrate to nitric oxide (NO) by purified CYP isoforms and the relationship between P450 expression and nitrate tolerance following continuous infusion of organic nitrates in rats. Nitric Oxide 73-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 12688525-7 2002 The appearance and disappearance of P450 isoforms paralleled the conversion of organic nitrates to NO as assessed by immunohistochemistry and Western blotting. Nitrates 87-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 12688525-8 2002 Our observations indicate that nitrate tolerance is in large part the result of decreased P450 expression and activity. Nitrates 31-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-94 11717183-0 2001 In vitro characterization of the metabolism of haloperidol using recombinant cytochrome p450 enzymes and human liver microsomes. Haloperidol 47-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 11804275-4 2001 This is achieved because CYP2B6 converts the inactive produg form cyclophosphamide into the active cytotoxic drug. Cyclophosphamide 66-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-31 11717183-1 2001 A systematic in vitro study was carried out to elucidate the enzymes responsible for the metabolism of haloperidol (HAL) using human liver microsomes and recombinant human cytochrome P450 isoenzymes. Haloperidol 103-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 11717183-1 2001 A systematic in vitro study was carried out to elucidate the enzymes responsible for the metabolism of haloperidol (HAL) using human liver microsomes and recombinant human cytochrome P450 isoenzymes. Haloperidol 116-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 11717183-2 2001 In the first series of experiments, recombinant cytochrome P450 (P450) isoenzymes were used to evaluate their catalytic involvement in the metabolic pathways of HAL. Haloperidol 161-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-63 11717183-2 2001 In the first series of experiments, recombinant cytochrome P450 (P450) isoenzymes were used to evaluate their catalytic involvement in the metabolic pathways of HAL. Haloperidol 161-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 11714868-6 2001 Rifampin also induced other CYP enzymes including CYP2B6 and all three members of the CYP3A family, with CYP3A4 showing the highest level of induction at 55.1-fold. Rifampin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 11875815-7 2001 RFP has a potency to induce the enzyme p450, which has the effect of metabolizing steroids or CyA, thus shortening the half-life of these agents. Steroids 82-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 11723234-2 2001 Tumor cells transduced with the human cytochrome P450 gene CYP2B6 are greatly sensitized to CPA, however, the pathway of CPA-induced cell death is unknown. Cyclophosphamide 92-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 11723234-2 2001 Tumor cells transduced with the human cytochrome P450 gene CYP2B6 are greatly sensitized to CPA, however, the pathway of CPA-induced cell death is unknown. Cyclophosphamide 121-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 11723234-3 2001 The present study investigates the cytotoxic events induced by CPA in 9L gliosarcoma cells retrovirally transduced with CYP2B6, or induced in wild-type 9L cells treated with mafosfamide (MFA) or 4-hydroperoxyifosfamide (4OOH-IFA), chemically activated forms of CPA and its isomer ifosfamide. Cyclophosphamide 63-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 11602510-6 2001 In a 2-min assay, purified 2D6 catalyzed the formation of dextrorphan with an apparent K(m) value of 1.9 +/- 0.2 microM and a V(max) value of 8.5 +/- 0.2 nmol/nmol of P450/min and measured simultaneously the formation of 3-methoxymorphinan with an apparent K(m) value of 5000 +/- 700 microM and V(max) value of 176 +/- 12 nmol (nmol of P450)(-1) min(-1). Tankyrase Inhibitors (TNKS) 49 27-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-175 11708902-3 2001 In an initial study using three human liver microsomal samples, K(m) for diazoxon formation varied markedly (31, 208, and 660 microM; V(max) 1125, 685, and 1028 pmol/min/mg protein, respectively), suggesting the involvement of more than one P450 enzyme. diazoxon 73-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 241-245 11708902-5 2001 Among eight P450-catalyzed reactions, the putative high-affinity component (50 microM diazinon) correlated with S-mephenytoin 4"-hydroxylase activity (r = 0.686, p < 0.01), suggesting the involvement of CYP2C19. Diazinon 86-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-16 11708902-10 2001 Of seven heterologously expressed human P450 enzymes, CYP2C19 activated diazinon (500 microM) at the fastest rate, followed by CYP3A4, CYP1A2, and CYP2C9. Diazinon 72-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-44 11602510-6 2001 In a 2-min assay, purified 2D6 catalyzed the formation of dextrorphan with an apparent K(m) value of 1.9 +/- 0.2 microM and a V(max) value of 8.5 +/- 0.2 nmol/nmol of P450/min and measured simultaneously the formation of 3-methoxymorphinan with an apparent K(m) value of 5000 +/- 700 microM and V(max) value of 176 +/- 12 nmol (nmol of P450)(-1) min(-1). Dextrorphan 58-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-175 11710930-7 2001 The ultraviolet-B-triggered conversion of 7-dehydrocholesterol to calcitriol was strongly inhibited by ketoconazole indicating the involvement of P450 mixed function oxidases. 7-dehydrocholesterol 42-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 146-150 11602525-0 2001 CYP2B6 and CYP2C19 as the major enzymes responsible for the metabolism of selegiline, a drug used in the treatment of Parkinson"s disease, as revealed from experiments with recombinant enzymes. Selegiline 74-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 11602525-3 2001 The highest contribution to the hepatic clearance of selegiline was calculated to be exerted by CYP2B6 (124 l/h) CYP2C19 (82 l/h), whereas CYP3A4 (27 l/h) and CYP1A2 (21 l/h) were of less importance. Selegiline 53-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 11602525-4 2001 Antibodies against CYP2B6 inhibited metabolism of selegiline in microsomes containing CYP2B6 but not in microsomes without significant amounts of the enzyme. Selegiline 50-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 11602525-4 2001 Antibodies against CYP2B6 inhibited metabolism of selegiline in microsomes containing CYP2B6 but not in microsomes without significant amounts of the enzyme. Selegiline 50-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 11602525-6 2001 The data strongly indicate that the high extent of interindividual variation seen in vivo for selegiline clearance is caused by the metabolism of the compound by the highly polymorphic CYP2B6 and CYP2C19. Selegiline 94-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 185-191 11710930-7 2001 The ultraviolet-B-triggered conversion of 7-dehydrocholesterol to calcitriol was strongly inhibited by ketoconazole indicating the involvement of P450 mixed function oxidases. Calcitriol 66-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 146-150 11710930-7 2001 The ultraviolet-B-triggered conversion of 7-dehydrocholesterol to calcitriol was strongly inhibited by ketoconazole indicating the involvement of P450 mixed function oxidases. Ketoconazole 103-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 146-150 11560876-8 2001 Finally, 10 microM CCM induced both CYP2C9 and CYP2B6, strengthening the evidence that hPXR is involved in the regulation of these genes. Fruitcal 19-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 11684365-6 2001 The formation of neoantigenic trifluoroacetylated protein adducts by microsomal mixtures incubated with HCFC-123 and NADPH was catalysed primarily by CYP2E1 and to a lesser extent by CYP2C19, whereas, only trace levels of immunoreactive protein were seen with microsomes expressing CYP2B6 or CYP2C8. NADP 117-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 282-288 11684365-8 2001 CYP1A2, CYP2B6 and CYP2C8 appeared to be responsible for perchloroethylene immunoreactivity and in contrast to the findings with the HCFC"s, no activation of perchloroethylene by CYP2E1 could be detected. Tetrachloroethylene 57-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 11560876-0 2001 Calcium channel modulators of the dihydropyridine family are human pregnane X receptor activators and inducers of CYP3A, CYP2B, and CYP2C in human hepatocytes. 1,4-dihydropyridine 34-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-126 11695850-0 2001 Roles of cytochrome P450 3A enzymes in the 2-hydroxylation of 1,4-cineole, a monoterpene cyclic ether, by rat and human liver microsomes. 1,4-cineole 62-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 11695850-0 2001 Roles of cytochrome P450 3A enzymes in the 2-hydroxylation of 1,4-cineole, a monoterpene cyclic ether, by rat and human liver microsomes. Monoterpenes 77-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 11695850-3 2001 On analysis with gas chromatography/mass spectrometry, 2-exo-hydroxy-1,4-cineole was identified as a principal oxidation product of 1,4-cineole catalysed by rat and human P450 enzymes. 2-Exo-hydroxy-1,4-cineole 55-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-175 11695850-0 2001 Roles of cytochrome P450 3A enzymes in the 2-hydroxylation of 1,4-cineole, a monoterpene cyclic ether, by rat and human liver microsomes. Ethers, Cyclic 89-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 11695850-3 2001 On analysis with gas chromatography/mass spectrometry, 2-exo-hydroxy-1,4-cineole was identified as a principal oxidation product of 1,4-cineole catalysed by rat and human P450 enzymes. 1,4-cineole 69-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-175 11518807-4 2001 Thus, the phenobarbital-inducible enhancer units of the mouse Cyp2b10, rat CYP2B2, and human CYP2B6 genes were activated in reporter gene assays by the same compounds that activate the chicken CYP2H1 phenobarbital-inducible enhancer units. Phenobarbital 10-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 11695850-8 2001 Finally, of 10 recombinant human P450 enzymes examined, CYP3A4 had the highest activity for 1,4-cineole 2-hydroxylation. 1,4-cineole 92-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 11502872-4 2001 In primary cultures of human hepatocytes, CYP2B6 was highly inducible by a number of compounds known to be human PXR ligands, including rifampicin and hyperforin. Rifampin 136-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 11502872-4 2001 In primary cultures of human hepatocytes, CYP2B6 was highly inducible by a number of compounds known to be human PXR ligands, including rifampicin and hyperforin. hyperforin 151-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 11502872-5 2001 PXR was shown to be capable of activating the phenobarbital-responsive enhancer module (PBREM) region of the CYP2B6 gene, a 51-base-pair enhancer element that mediates induction of CYP2B6 expression by CAR. Phenobarbital 46-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 11502872-5 2001 PXR was shown to be capable of activating the phenobarbital-responsive enhancer module (PBREM) region of the CYP2B6 gene, a 51-base-pair enhancer element that mediates induction of CYP2B6 expression by CAR. Phenobarbital 46-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-187 11518807-4 2001 Thus, the phenobarbital-inducible enhancer units of the mouse Cyp2b10, rat CYP2B2, and human CYP2B6 genes were activated in reporter gene assays by the same compounds that activate the chicken CYP2H1 phenobarbital-inducible enhancer units. Phenobarbital 200-213 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 11469860-1 2001 The azole-based P450 inhibitor ketoconazole is used to treat fungal infections and functions by blocking ergosterol biosynthesis in yeast. Azoles 4-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 11513592-2 2001 The sites were chosen on the basis of previous studies or from a comparison with the structure of P450(eryF) containing two molecules of androstenedione. Androstenedione 137-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-108 11469860-1 2001 The azole-based P450 inhibitor ketoconazole is used to treat fungal infections and functions by blocking ergosterol biosynthesis in yeast. Ketoconazole 31-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 11469860-1 2001 The azole-based P450 inhibitor ketoconazole is used to treat fungal infections and functions by blocking ergosterol biosynthesis in yeast. Ergosterol 105-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 11469860-2 2001 Ketoconazole binds to mammalian P450 enzymes and this can result in drug-drug interactions and lead to liver damage. Ketoconazole 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 11503007-3 2001 The aim of this study was to investigate modulation of the CYP-mediated metabolism of ifosfamide with ketoconazole, a potent inhibitor of CYP3A4, and rifampin (INN, rifampicin), an inducer of CYP3A4/CYP2B6. Rifampin 150-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-205 11454734-7 2001 In studies with characterized human liver microsomal preparations, good correlations were observed between tramadol metabolism to M1 and M2 and enzymatic markers of CYP2D6 and CYP2B6, respectively. Tramadol 107-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 176-182 11503007-3 2001 The aim of this study was to investigate modulation of the CYP-mediated metabolism of ifosfamide with ketoconazole, a potent inhibitor of CYP3A4, and rifampin (INN, rifampicin), an inducer of CYP3A4/CYP2B6. Ifosfamide 86-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-205 11503007-3 2001 The aim of this study was to investigate modulation of the CYP-mediated metabolism of ifosfamide with ketoconazole, a potent inhibitor of CYP3A4, and rifampin (INN, rifampicin), an inducer of CYP3A4/CYP2B6. Ketoconazole 102-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-205 11454734-8 2001 Tramadol was metabolized to M1 by cDNA-expressed CYP2D6 and to M2 by CYP2B6 and CYP3A4. Tramadol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 11498768-1 2001 Human cytochrome P450 2B6 (CYP2B6) metabolizes the prodrug cyclophosphamide (CPA) to produce phosphoramide mustard that cross-links DNA leading to cell death. Cyclophosphamide 59-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-25 11485379-1 2001 Ketoximes undergo a cytochrome P450-catalyzed oxidation to nitric oxide and ketones in liver microsomes. Nitric Oxide 59-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 11485379-1 2001 Ketoximes undergo a cytochrome P450-catalyzed oxidation to nitric oxide and ketones in liver microsomes. Ketones 76-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 11485379-4 2001 Acetoxime was oxidized to NO2- (and NO3-) by microsomes enriched with several P450 isoforms, including CYP2E1, CYP1A1, and CYP2B1. acetoxime 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 11485379-4 2001 Acetoxime was oxidized to NO2- (and NO3-) by microsomes enriched with several P450 isoforms, including CYP2E1, CYP1A1, and CYP2B1. Nitrogen Dioxide 26-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 11498768-1 2001 Human cytochrome P450 2B6 (CYP2B6) metabolizes the prodrug cyclophosphamide (CPA) to produce phosphoramide mustard that cross-links DNA leading to cell death. Cyclophosphamide 77-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-25 11498768-1 2001 Human cytochrome P450 2B6 (CYP2B6) metabolizes the prodrug cyclophosphamide (CPA) to produce phosphoramide mustard that cross-links DNA leading to cell death. Cyclophosphamide 77-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 11498768-1 2001 Human cytochrome P450 2B6 (CYP2B6) metabolizes the prodrug cyclophosphamide (CPA) to produce phosphoramide mustard that cross-links DNA leading to cell death. Cyclophosphamide 59-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 11498768-1 2001 Human cytochrome P450 2B6 (CYP2B6) metabolizes the prodrug cyclophosphamide (CPA) to produce phosphoramide mustard that cross-links DNA leading to cell death. phosphoramide mustard 93-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-25 11412116-5 2001 Studies of P450 11A1 indicate that Phe-202 has functions similar to those of its counterpart in P450 27A1 (Phe-215). Phenylalanine 35-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 11498768-1 2001 Human cytochrome P450 2B6 (CYP2B6) metabolizes the prodrug cyclophosphamide (CPA) to produce phosphoramide mustard that cross-links DNA leading to cell death. phosphoramide mustard 93-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 11416012-2 2001 low density lipoprotein (LDL) receptor, steroidogenic acute regulatory protein, and P450 cholesterol side-chain cleavage enzyme (CYP11A). Cholesterol 89-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 11412116-5 2001 Studies of P450 11A1 indicate that Phe-202 has functions similar to those of its counterpart in P450 27A1 (Phe-215). Phenylalanine 107-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-100 11396966-0 2001 Differential N-demethylation of l-alpha-acetylmethadol (LAAM) and norLAAM by cytochrome P450s 2B6, 2C18, and 3A4. Nitrogen 13-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-103 11396966-0 2001 Differential N-demethylation of l-alpha-acetylmethadol (LAAM) and norLAAM by cytochrome P450s 2B6, 2C18, and 3A4. Methadyl Acetate 32-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-103 11396966-0 2001 Differential N-demethylation of l-alpha-acetylmethadol (LAAM) and norLAAM by cytochrome P450s 2B6, 2C18, and 3A4. Methadyl Acetate 56-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-103 11396966-0 2001 Differential N-demethylation of l-alpha-acetylmethadol (LAAM) and norLAAM by cytochrome P450s 2B6, 2C18, and 3A4. paracymethadol 66-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-103 11469727-1 2001 Cytochrome P450 (P450) enzymes play major roles in the metabolism of drugs, carcinogens, steroids, eicosanoids, alkaloids, pesticides, and other important xenobiotics, as well as chemicals normally endogenous to the body. Steroids 89-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 11469727-1 2001 Cytochrome P450 (P450) enzymes play major roles in the metabolism of drugs, carcinogens, steroids, eicosanoids, alkaloids, pesticides, and other important xenobiotics, as well as chemicals normally endogenous to the body. Eicosanoids 99-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 11469727-1 2001 Cytochrome P450 (P450) enzymes play major roles in the metabolism of drugs, carcinogens, steroids, eicosanoids, alkaloids, pesticides, and other important xenobiotics, as well as chemicals normally endogenous to the body. Alkaloids 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-21 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 2 subfamily B member 6 Homo sapiens 13-17 11353747-0 2001 A novel cytochrome P450 enzyme responsible for the metabolism of ebastine in monkey small intestine. ebastine 65-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 11353747-2 2001 To identify the main enzyme responsible for ebastine hydroxylation, which has been hitherto unknown, we purified two cytochrome P450 isoforms, named P450 MI-2 and P450 MI-3, from the intestinal microsomes on the basis of the hydroxylation activity. ebastine 44-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 11353747-2 2001 To identify the main enzyme responsible for ebastine hydroxylation, which has been hitherto unknown, we purified two cytochrome P450 isoforms, named P450 MI-2 and P450 MI-3, from the intestinal microsomes on the basis of the hydroxylation activity. ebastine 44-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-153 11353747-2 2001 To identify the main enzyme responsible for ebastine hydroxylation, which has been hitherto unknown, we purified two cytochrome P450 isoforms, named P450 MI-2 and P450 MI-3, from the intestinal microsomes on the basis of the hydroxylation activity. ebastine 44-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-153 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 2 subfamily B member 6 Homo sapiens 27-31 11353747-3 2001 P450 MI-2 and P450 MI-3 showed the respective apparent molecular weights of 56,000 and 53,000 on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Sodium Dodecyl Sulfate 97-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 11353747-3 2001 P450 MI-2 and P450 MI-3 showed the respective apparent molecular weights of 56,000 and 53,000 on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Sodium Dodecyl Sulfate 97-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 11353747-3 2001 P450 MI-2 and P450 MI-3 showed the respective apparent molecular weights of 56,000 and 53,000 on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. polyacrylamide 120-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 11353747-3 2001 P450 MI-2 and P450 MI-3 showed the respective apparent molecular weights of 56,000 and 53,000 on sodium dodecyl sulfate-polyacrylamide gel electrophoresis. polyacrylamide 120-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 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 2 subfamily B member 6 Homo sapiens 13-17 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 2 subfamily B member 6 Homo sapiens 27-31 11353747-7 2001 Although both enzymes had ebastine hydroxylation activity, the V(max) value of P450 MI-2 was much higher than that of P450 MI-3 (37.0 versus 0.406 nmol/min/nmol of P450), and the former K(M) (5.1 microM) was smaller than the latter K(M) (10 microM). ebastine 26-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 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 2 subfamily B member 6 Homo sapiens 28-32 11259570-7 2001 Of the cDNA-expressed isoforms examined, CYP2B6 and 3A4 had the highest activity toward LAAM and nor-LAAM at both low (2 microM) and high (250 microM) substrate concentrations. Methadyl Acetate 88-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 11353758-0 2001 Involvement of CYP2B6 in n-demethylation of ketamine in human liver microsomes. Nitrogen 1-2 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 11353758-0 2001 Involvement of CYP2B6 in n-demethylation of ketamine in human liver microsomes. Ketamine 44-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 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 2 subfamily B member 6 Homo sapiens 50-56 11353758-6 2001 CYP2B6 had the lowest K(m) value for the N-demethylation of (R)- and (S)-ketamine (74 and 44 microM, respectively). Nitrogen 41-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 11353758-6 2001 CYP2B6 had the lowest K(m) value for the N-demethylation of (R)- and (S)-ketamine (74 and 44 microM, respectively). Ranolazine 60-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 11353758-6 2001 CYP2B6 had the lowest K(m) value for the N-demethylation of (R)- and (S)-ketamine (74 and 44 microM, respectively). Ketamine 69-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 11353758-7 2001 Also, the intrinsic clearance (CL(int): V(max)/K(m)) of CYP2B6 for the N-demethylation of both enantiomers were 7 to 13 times higher than those of CYP2C9 and CYP3A4. Nitrogen 71-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 11353758-8 2001 Orphenadrine (CYP2B6 inhibitor, 500 microM) and sulfaphenazole (CYP2C9 inhibitor, 100 microM) inhibited the N-demethylase activities for both enantiomers (5 microM) in human liver microsomes by 60 to 70%, whereas cyclosporin A (CYP3A4 inhibitor, 100 microM) failed to inhibit these activities. Orphenadrine 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 11353758-11 2001 CYP2B6 mainly mediates the N-demethylation of (R)- and (S)-ketamine in human liver microsomes at therapeutic concentrations (5 microM). Nitrogen 27-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 11353758-11 2001 CYP2B6 mainly mediates the N-demethylation of (R)- and (S)-ketamine in human liver microsomes at therapeutic concentrations (5 microM). (r)- and (s)-ketamine 46-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 11356927-0 2001 Identification of the human cytochromes p450 responsible for in vitro formation of R- and S-norfluoxetine. r- and s-norfluoxetine 83-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-44 11377247-0 2001 Metabolic activation of heterocyclic amines and other procarcinogens in Salmonella typhimurium umu tester strains expressing human cytochrome P4501A1, 1A2, 1B1, 2C9, 2D6, 2E1, and 3A4 and human NADPH-P450 reductase and bacterial O-acetyltransferase. heterocyclic amines 24-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 11573629-4 2001 Midazolam 1"-hydroxylation activity of human liver microsomes correlated significantly with testosterone 6beta-hydroxylation activity, a marker of CYP3A activity (r2 = 0.77, P = 0.0001), but not with S-mephenytoin N-demethylation activity, a marker of CYP2B6 activity (r2 < 0.01, P = 0.84) (n = 11). Midazolam 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 252-258 11573629-7 2001 These results indicate that only CYP3A4, but not CYP2B6 or CYP2C, is involved in the metabolism of midazolam in vitro. Midazolam 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 11259560-4 2001 For the pathways of N-demethylation (mediated by CYPs 1A2, 2B6, 2C8, 2C9, 2C19, 2D6, and 3A4) and E-10 hydroxylation (mediated by CYPs 2B6, 2D6, and 3A4), the model-predicted biotransformation rates in microsomes from a panel of 12 human livers determined from enzyme kinetic parameters of the recombinant CYPs were similar to, and correlated with the observed rates. Nitrogen 20-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-143 11259570-7 2001 Of the cDNA-expressed isoforms examined, CYP2B6 and 3A4 had the highest activity toward LAAM and nor-LAAM at both low (2 microM) and high (250 microM) substrate concentrations. paracymethadol 97-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 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 2 subfamily B member 6 Homo sapiens 123-129 11257517-7 2001 P450 enzyme activities were assessed using 8 different substrates and increases were found after treatment with phenobarbitone, rifampicin, and dioxin. Phenobarbital 112-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 11257517-7 2001 P450 enzyme activities were assessed using 8 different substrates and increases were found after treatment with phenobarbitone, rifampicin, and dioxin. Rifampin 128-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 11257517-7 2001 P450 enzyme activities were assessed using 8 different substrates and increases were found after treatment with phenobarbitone, rifampicin, and dioxin. Dioxins 144-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 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 2 subfamily B member 6 Homo sapiens 67-73 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 2 subfamily B member 6 Homo sapiens 67-73 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 2 subfamily B member 6 Homo sapiens 42-48 11298076-8 2001 An antibody against CYP2B6 inhibited the disappearance of propofol in liver microsomes by 74%. Propofol 58-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 11298076-10 2001 CONCLUSIONS: CYP2B6 is predominantly involved in the oxidation of propofol by human liver microsomes. Propofol 66-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 11181500-2 2001 All 60 tumor cell lines displayed significant P450 activity, as well as P450 reductase activity, as determined using the general P450 substrate 7-benzyloxyresorufin. benzyloxyresorufin 144-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-76 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 71-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 11181500-2 2001 All 60 tumor cell lines displayed significant P450 activity, as well as P450 reductase activity, as determined using the general P450 substrate 7-benzyloxyresorufin. benzyloxyresorufin 144-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-76 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Phenobarbital 86-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 192-202 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 11181500-3 2001 Cell line-specific P450 enzyme patterns were observed using three other P450 substrates, 7-ethoxycoumarin, coumarin, and 7-ethoxyresorufin, each of which was metabolized at a low rate. 7-ethoxycoumarin 89-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Phenobarbital 218-231 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 11181500-3 2001 Cell line-specific P450 enzyme patterns were observed using three other P450 substrates, 7-ethoxycoumarin, coumarin, and 7-ethoxyresorufin, each of which was metabolized at a low rate. coumarin 97-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 11181500-3 2001 Cell line-specific P450 enzyme patterns were observed using three other P450 substrates, 7-ethoxycoumarin, coumarin, and 7-ethoxyresorufin, each of which was metabolized at a low rate. ethoxyresorufin 121-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 11181500-5 2001 Significant negative correlations between the patterns of P450-dependent 7-benzyloxyresorufin metabolism activity and cell line chemosensitivity were observed for 10 standard anticancer agents (including 6 alkylating agents) and 55 investigational compounds, suggesting a role for P450 metabolism in the inactivation of these agents. benzyloxyresorufin 73-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 281-285 11181500-5 2001 Significant negative correlations between the patterns of P450-dependent 7-benzyloxyresorufin metabolism activity and cell line chemosensitivity were observed for 10 standard anticancer agents (including 6 alkylating agents) and 55 investigational compounds, suggesting a role for P450 metabolism in the inactivation of these agents. benzyloxyresorufin 73-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 11181500-6 2001 Negative correlations between 7-ethoxycoumarin O-deethylation and cell line chemosensitivity to a group of topoisomerase inhibitors were also seen, again suggesting P450-dependent drug inactivation. 7-ethoxycoumarin 30-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-169 11231298-5 2001 Methylcholanthrene induced an increase in CYP1A1/2 enzyme activity (eightfold), phenobarbital induced CYP2B6 activity (1.7-fold), and dexamethasone induced CYP3A4 activity (fivefold). Phenobarbital 80-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 11159809-13 2001 Human CYP2B6 preferentially formed VCH-7,8-epoxide compared with VCH-1,2-epoxide, and to a greater extent from (R)-VCH than from (S)-VCH. vch-7,8-epoxide 35-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 11411236-1 2001 In this paper, we studied the effects of the adding time of alkane on the expression of P450 and production of dicarboxylic acid. Alkanes 60-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 11411236-2 2001 A novel fermentation process, in which no or a little alkane was added to make the cells growing more quickly during the growth stage, followed by the addition of alkane to induce cytochromes P450 for 6-8 hours, was established. Alkanes 163-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-196 11159797-0 2001 Ritonavir, efavirenz, and nelfinavir inhibit CYP2B6 activity in vitro: potential drug interactions with bupropion. Ritonavir 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 11159797-0 2001 Ritonavir, efavirenz, and nelfinavir inhibit CYP2B6 activity in vitro: potential drug interactions with bupropion. efavirenz 11-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 11159797-0 2001 Ritonavir, efavirenz, and nelfinavir inhibit CYP2B6 activity in vitro: potential drug interactions with bupropion. Nelfinavir 26-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 11159797-0 2001 Ritonavir, efavirenz, and nelfinavir inhibit CYP2B6 activity in vitro: potential drug interactions with bupropion. Bupropion 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 11159809-13 2001 Human CYP2B6 preferentially formed VCH-7,8-epoxide compared with VCH-1,2-epoxide, and to a greater extent from (R)-VCH than from (S)-VCH. vch-1,2-epoxide 65-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 11159797-4 2001 The inhibition of bupropion hydroxylation in vitro by nelfinavir, ritonavir, and efavirenz indicates inhibitory potency versus CYP2B6 and suggests the potential for clinical drug interactions. Bupropion 18-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 11159797-4 2001 The inhibition of bupropion hydroxylation in vitro by nelfinavir, ritonavir, and efavirenz indicates inhibitory potency versus CYP2B6 and suggests the potential for clinical drug interactions. Nelfinavir 54-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 11159809-13 2001 Human CYP2B6 preferentially formed VCH-7,8-epoxide compared with VCH-1,2-epoxide, and to a greater extent from (R)-VCH than from (S)-VCH. (r)-vch 111-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 11159812-2 2001 We investigated the metabolism of 1,8-cineole by liver microsomes of rats and humans and by recombinant cytochrome P450 (P450 or CYP) enzymes in insect cells in which human P450 and NADPH-P450 reductase cDNAs had been introduced. Eucalyptol 34-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-132 11159812-2 2001 We investigated the metabolism of 1,8-cineole by liver microsomes of rats and humans and by recombinant cytochrome P450 (P450 or CYP) enzymes in insect cells in which human P450 and NADPH-P450 reductase cDNAs had been introduced. Eucalyptol 34-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 11159809-13 2001 Human CYP2B6 preferentially formed VCH-7,8-epoxide compared with VCH-1,2-epoxide, and to a greater extent from (R)-VCH than from (S)-VCH. 4-vinylcyclohexene 35-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 11159812-2 2001 We investigated the metabolism of 1,8-cineole by liver microsomes of rats and humans and by recombinant cytochrome P450 (P450 or CYP) enzymes in insect cells in which human P450 and NADPH-P450 reductase cDNAs had been introduced. Eucalyptol 34-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 11159812-3 2001 1,8-Cineole was found to be oxidized at high rates to 2-exo-hydroxy-1,8-cineole by rat and human liver microsomal P450 enzymes. Eucalyptol 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-118 11159812-3 2001 1,8-Cineole was found to be oxidized at high rates to 2-exo-hydroxy-1,8-cineole by rat and human liver microsomal P450 enzymes. 6-endo-Hydroxycineole 54-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-118 11159812-0 2001 Oxidation of 1,8-cineole, the monoterpene cyclic ether originated from eucalyptus polybractea, by cytochrome P450 3A enzymes in rat and human liver microsomes. Eucalyptol 13-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 11159812-6 2001 Kinetic analysis showed that K(m) and V(max) values for the oxidation of 1,8-cineole by liver microsomes of human sample HL-104 and rats treated with PCN were 50 microM and 91 nmol/min/nmol P450 and 20 microM and 12 nmol/min/nmol P450, respectively. Eucalyptol 73-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 190-194 11159812-6 2001 Kinetic analysis showed that K(m) and V(max) values for the oxidation of 1,8-cineole by liver microsomes of human sample HL-104 and rats treated with PCN were 50 microM and 91 nmol/min/nmol P450 and 20 microM and 12 nmol/min/nmol P450, respectively. Eucalyptol 73-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 230-234 11159812-6 2001 Kinetic analysis showed that K(m) and V(max) values for the oxidation of 1,8-cineole by liver microsomes of human sample HL-104 and rats treated with PCN were 50 microM and 91 nmol/min/nmol P450 and 20 microM and 12 nmol/min/nmol P450, respectively. PREGNENOLONE CARBONITRILE 150-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 190-194 11159812-6 2001 Kinetic analysis showed that K(m) and V(max) values for the oxidation of 1,8-cineole by liver microsomes of human sample HL-104 and rats treated with PCN were 50 microM and 91 nmol/min/nmol P450 and 20 microM and 12 nmol/min/nmol P450, respectively. PREGNENOLONE CARBONITRILE 150-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 230-234 11159812-0 2001 Oxidation of 1,8-cineole, the monoterpene cyclic ether originated from eucalyptus polybractea, by cytochrome P450 3A enzymes in rat and human liver microsomes. Monoterpenes 30-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 11159812-0 2001 Oxidation of 1,8-cineole, the monoterpene cyclic ether originated from eucalyptus polybractea, by cytochrome P450 3A enzymes in rat and human liver microsomes. Ethers, Cyclic 42-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 11159812-2 2001 We investigated the metabolism of 1,8-cineole by liver microsomes of rats and humans and by recombinant cytochrome P450 (P450 or CYP) enzymes in insect cells in which human P450 and NADPH-P450 reductase cDNAs had been introduced. Eucalyptol 34-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-119 11240372-2 2001 Sodium cholate and mercury chloride induced the conversion of nNOS from the P-450 to the P-420 form in concentration- and incubation time-dependent manners, and the nNOS activity decreased. Sodium Cholate 0-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-81 11240372-2 2001 Sodium cholate and mercury chloride induced the conversion of nNOS from the P-450 to the P-420 form in concentration- and incubation time-dependent manners, and the nNOS activity decreased. calomel 19-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-81 11240372-0 2001 The optical interconversion of the P-450 and P-420 forms of neuronal nitric oxide synthase: effects of sodium cholate, mercury chloride and urea. Sodium Cholate 103-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-90 11240372-3 2001 In the presence of glycerol, L-arginine and/or tetrahydrobiopterin, the sodium cholate-treated P-420 form could be reconverted to the P-450 form under constant experimental conditions, and the nNOS activity could also be restored. Glycerol 19-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 11240372-0 2001 The optical interconversion of the P-450 and P-420 forms of neuronal nitric oxide synthase: effects of sodium cholate, mercury chloride and urea. calomel 119-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-90 11240372-0 2001 The optical interconversion of the P-450 and P-420 forms of neuronal nitric oxide synthase: effects of sodium cholate, mercury chloride and urea. Urea 140-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-90 11240372-3 2001 In the presence of glycerol, L-arginine and/or tetrahydrobiopterin, the sodium cholate-treated P-420 form could be reconverted to the P-450 form under constant experimental conditions, and the nNOS activity could also be restored. Arginine 29-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 11240372-3 2001 In the presence of glycerol, L-arginine and/or tetrahydrobiopterin, the sodium cholate-treated P-420 form could be reconverted to the P-450 form under constant experimental conditions, and the nNOS activity could also be restored. sapropterin 47-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 11240372-3 2001 In the presence of glycerol, L-arginine and/or tetrahydrobiopterin, the sodium cholate-treated P-420 form could be reconverted to the P-450 form under constant experimental conditions, and the nNOS activity could also be restored. Sodium Cholate 72-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 11240372-4 2001 The mercury chloride-treated P-420 form of nNOS could be reconverted to the P-450 form on incubation with reduced glutathione (GSH) or L-cysteine, and the nNOS activity was recovered. calomel 4-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-81 11240372-4 2001 The mercury chloride-treated P-420 form of nNOS could be reconverted to the P-450 form on incubation with reduced glutathione (GSH) or L-cysteine, and the nNOS activity was recovered. Glutathione 114-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-81 11240372-4 2001 The mercury chloride-treated P-420 form of nNOS could be reconverted to the P-450 form on incubation with reduced glutathione (GSH) or L-cysteine, and the nNOS activity was recovered. Glutathione 127-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-81 11240372-4 2001 The mercury chloride-treated P-420 form of nNOS could be reconverted to the P-450 form on incubation with reduced glutathione (GSH) or L-cysteine, and the nNOS activity was recovered. Cysteine 135-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-81 11135730-0 2001 Cytochrome P-450 2B6 is responsible for interindividual variability of propofol hydroxylation by human liver microsomes. Propofol 71-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-20 11135730-6 2001 RESULTS: Propofol hydroxylation by hepatic microsomes showed more than 19-fold variability and was most closely correlated to CYP2B6 protein content (r = 0.904), and the CYP2B6 marker activities, S-mephenytoin N-demethylation (r = 0.919) and bupropion hydroxylation (r = 0.854). Propofol 9-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 11135730-6 2001 RESULTS: Propofol hydroxylation by hepatic microsomes showed more than 19-fold variability and was most closely correlated to CYP2B6 protein content (r = 0.904), and the CYP2B6 marker activities, S-mephenytoin N-demethylation (r = 0.919) and bupropion hydroxylation (r = 0.854). Propofol 9-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-176 11135730-8 2001 All of the CYPs evaluated were capable of hydroxylating propofol; however, CYP2B6 and CYP2C9 were most active. Propofol 56-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 11135730-11 2001 CONCLUSIONS: Cytochrome P-450 2B6, and to a lesser extent CYP2C9, contribute to the oxidative metabolism of propofol. Propofol 108-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-33 11554431-0 2001 Interactions of melatonin with the liver microsomal cytochrome P450 system of rats and humans in vitro and effects on the P450 system and the antioxidative status in rat liver after acute treatment. Melatonin 16-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 11432537-10 2001 CYP2B6 and CYP3A4 are involved in the metabolism of artemisinin and derivatives, but further studies may reveal involvement of more enzymes. artemisinin 52-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 11554431-1 2001 In vitro melatonin binds to human and rat liver microsomal cytochrome P-450 (P450) according to a type II substrate. Melatonin 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-81 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. Melatonin 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. Melatonin 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. Melatonin 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. Melatonin 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. Melatonin 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. ethoxyresorufin 223-238 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. ethoxyresorufin 223-238 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. ethoxyresorufin 223-238 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. ethoxyresorufin 223-238 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 11554431-3 2001 Melatonin interferes with model monooxygenase reactions indicative of different P450 forms in humans and rats (in humans according to the lower specific P450 content less pronounced): the strongest inhibition was found for ethoxyresorufin O-deethylation, indicating the binding to P450 1A, the binding to P450 2B (ethoxycoumarin O-deethylation) was less pronounced, the least inhibition was found for P450 3A (ethylmorphine N-demethylation) reaction. ethoxyresorufin 223-238 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 11908754-1 2001 P450 enzymes comprise a superfamily of heme-containing proteins that catalyze oxidative metabolism of structurally diverse chemicals. Heme 39-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 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 2 subfamily B member 6 Homo sapiens 653-659 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 2 subfamily B member 6 Homo sapiens 653-659 11095583-0 2000 Roles of cytochromes P450 1A2, 2A6, and 2C8 in 5-fluorouracil formation from tegafur, an anticancer prodrug, in human liver microsomes. Fluorouracil 47-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 11133395-10 2000 We have also shown that the cytochrome P450 isoforms responsible for human metabolism of acetochlor, butachlor, and metolachlor are CYP3A4 and CYP2B6. acetochlor 89-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-149 11095583-1 2000 Tegafur, an anticancer prodrug, is bioactivated to 5-fluorouracil (5-FU) mainly by cytochrome P450 (P450) enzymes. Tegafur 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-98 11095583-1 2000 Tegafur, an anticancer prodrug, is bioactivated to 5-fluorouracil (5-FU) mainly by cytochrome P450 (P450) enzymes. Tegafur 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-104 11095583-1 2000 Tegafur, an anticancer prodrug, is bioactivated to 5-fluorouracil (5-FU) mainly by cytochrome P450 (P450) enzymes. Fluorouracil 51-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-98 11095583-1 2000 Tegafur, an anticancer prodrug, is bioactivated to 5-fluorouracil (5-FU) mainly by cytochrome P450 (P450) enzymes. Fluorouracil 51-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-104 11095583-1 2000 Tegafur, an anticancer prodrug, is bioactivated to 5-fluorouracil (5-FU) mainly by cytochrome P450 (P450) enzymes. Fluorouracil 67-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-98 11095583-1 2000 Tegafur, an anticancer prodrug, is bioactivated to 5-fluorouracil (5-FU) mainly by cytochrome P450 (P450) enzymes. Fluorouracil 67-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-104 11095583-2 2000 The conversion from tegafur into 5-FU catalyzed by human liver microsomal P450 enzymes was investigated. Tegafur 20-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-78 11095583-2 2000 The conversion from tegafur into 5-FU catalyzed by human liver microsomal P450 enzymes was investigated. Fluorouracil 33-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-78 11095583-3 2000 In fourteen cDNA-expressed human P450 enzymes having measurable activities, CYP1A2, CYP2A6, CYP2E1, and CYP3A5 were highly active in catalyzing 5-FU formation at a tegafur concentration of 100 microM. Fluorouracil 144-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 11095583-10 2000 These results suggest that CYP1A2, CYP2A6, and CYP2C8 have important roles in human liver microsomal 5-FU formation and that the involvement of these three P450 forms differs among individual humans. Fluorouracil 101-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 156-160 11093772-1 2000 Cytochrome P450 (P450)-dependent metabolism of all-trans-retinoic acid (atRA) is important for the expression of its biological activity. Tretinoin 47-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 11093772-1 2000 Cytochrome P450 (P450)-dependent metabolism of all-trans-retinoic acid (atRA) is important for the expression of its biological activity. Tretinoin 72-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 11093784-4 2000 We show that submicromolar concentrations of dexamethasone enhance phenobarbital-mediated induction of CYP3A4, CYP2B6, and CYP2C8 mRNA in cultured human hepatocytes. Phenobarbital 67-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 11093784-2 2000 CAR is a member of the nuclear receptor family (NR1) mostly expressed in the liver, which heterodimerizes with retinoid X receptor (RXR) and was shown to transactivate both the phenobarbital responsive element module of the human CYP2B6 gene and the CYP3A4 xenobiotic response element. Phenobarbital 177-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 230-236 11038165-9 2000 These results suggest that CYP2C9, CYP2C19, and CYP3A4 all have catalytic activities in 3",4"-diHPPH formation from primary hydroxylated metabolites in human liver and that the hepatic contents of these three P450 forms determine which P450 enzymes play major roles of DPH oxidation in individual humans. 3",4"-dihpph 88-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 209-213 11093784-4 2000 We show that submicromolar concentrations of dexamethasone enhance phenobarbital-mediated induction of CYP3A4, CYP2B6, and CYP2C8 mRNA in cultured human hepatocytes. Dexamethasone 45-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 11140269-6 2000 The conversion of vitamin D3 generated after UVB irradiation to calcitriol is inhibited by ketoconazole indicating the involvement of P450 mixed function oxidases in this chemical reaction. Cholecalciferol 18-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-138 11140269-6 2000 The conversion of vitamin D3 generated after UVB irradiation to calcitriol is inhibited by ketoconazole indicating the involvement of P450 mixed function oxidases in this chemical reaction. Calcitriol 64-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-138 11140269-6 2000 The conversion of vitamin D3 generated after UVB irradiation to calcitriol is inhibited by ketoconazole indicating the involvement of P450 mixed function oxidases in this chemical reaction. Ketoconazole 91-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-138 11097178-0 2000 Structural similarities and differences of the heme pockets of various P450 isoforms as revealed by resonance Raman spectroscopy. Heme 47-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 11097178-7 2000 The spectral differences associated with the porphyrin substituents are likely to reflect subtle conformational differences in the heme pocket of various P450 isoforms which may constitute the structural basis for the known variability of their functions. Heme 131-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 11038165-1 2000 Formation of four oxidative metabolites from the anticonvulsant drug phenytoin (DPH) catalyzed by human liver microsomal cytochrome P450 (P450) enzymes was determined simultaneously. Phenytoin 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-136 11038165-1 2000 Formation of four oxidative metabolites from the anticonvulsant drug phenytoin (DPH) catalyzed by human liver microsomal cytochrome P450 (P450) enzymes was determined simultaneously. Phenytoin 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-142 11038165-1 2000 Formation of four oxidative metabolites from the anticonvulsant drug phenytoin (DPH) catalyzed by human liver microsomal cytochrome P450 (P450) enzymes was determined simultaneously. Phenytoin 80-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-136 11038165-9 2000 These results suggest that CYP2C9, CYP2C19, and CYP3A4 all have catalytic activities in 3",4"-diHPPH formation from primary hydroxylated metabolites in human liver and that the hepatic contents of these three P450 forms determine which P450 enzymes play major roles of DPH oxidation in individual humans. 3",4"-dihpph 88-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 236-240 11038165-1 2000 Formation of four oxidative metabolites from the anticonvulsant drug phenytoin (DPH) catalyzed by human liver microsomal cytochrome P450 (P450) enzymes was determined simultaneously. Phenytoin 80-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-142 11038165-7 2000 Of 10 cDNA-expressed human P450 enzymes examined, CYP2C19, CYP2C9, and CYP3A4 catalyzed 3",4"-diHPPH formation from the primary hydroxylated metabolites (3"-hydroxy-DPH and 4"-HPPH). 3",4"-dihpph 88-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 11027970-0 2000 Plant activation of aromatic amines mediated by cytochromes P450 and flavin-containing monooxygenases. aromatic amines 20-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-64 11038165-7 2000 Of 10 cDNA-expressed human P450 enzymes examined, CYP2C19, CYP2C9, and CYP3A4 catalyzed 3",4"-diHPPH formation from the primary hydroxylated metabolites (3"-hydroxy-DPH and 4"-HPPH). 3"-hydroxy-dph 154-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 11038165-7 2000 Of 10 cDNA-expressed human P450 enzymes examined, CYP2C19, CYP2C9, and CYP3A4 catalyzed 3",4"-diHPPH formation from the primary hydroxylated metabolites (3"-hydroxy-DPH and 4"-HPPH). hydroxyphenytoin 173-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 11132619-0 2000 A new thioether-ligated iron porphyrin as a model of a protonated form of P450 active site. Sulfides 6-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-78 11132619-0 2000 A new thioether-ligated iron porphyrin as a model of a protonated form of P450 active site. iron porphyrin 24-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-78 11132619-1 2000 Thioether-ligated iron porphyrin (complex 1) was synthesized as a model of the protonated form of P450 to explore the possible involvement of the protonated form in the catalytic cycle, and ether-ligated iron porphyrin (complex 2) was also synthesized for comparison. Sulfides 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 11132619-1 2000 Thioether-ligated iron porphyrin (complex 1) was synthesized as a model of the protonated form of P450 to explore the possible involvement of the protonated form in the catalytic cycle, and ether-ligated iron porphyrin (complex 2) was also synthesized for comparison. iron porphyrin 18-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 11132619-3 2000 Competitive oxidation of cyclooctane/cyclooctene catalyzed by iron porphyrins showed that complexes 1 and 2 are less effective than heme thiolate (P450 and a synthetic heme thiolate (SR complex)) in oxidizing alkane. cyclooctane 25-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-151 11132619-3 2000 Competitive oxidation of cyclooctane/cyclooctene catalyzed by iron porphyrins showed that complexes 1 and 2 are less effective than heme thiolate (P450 and a synthetic heme thiolate (SR complex)) in oxidizing alkane. Cyclooctanes 37-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-151 11132619-3 2000 Competitive oxidation of cyclooctane/cyclooctene catalyzed by iron porphyrins showed that complexes 1 and 2 are less effective than heme thiolate (P450 and a synthetic heme thiolate (SR complex)) in oxidizing alkane. iron porphyrins 62-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-151 11132619-3 2000 Competitive oxidation of cyclooctane/cyclooctene catalyzed by iron porphyrins showed that complexes 1 and 2 are less effective than heme thiolate (P450 and a synthetic heme thiolate (SR complex)) in oxidizing alkane. heme thiolate 132-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-151 11027970-4 2000 Similarly, both inhibitors and methimazole did produce a significant decrease in 2-AF and m-PDA mutagenesis, when the activation system was S117, indicating that, similar to what occurs in mammalian systems, plant FMOs and cyt-P450s can metabolize aromatic amines to mutagenic product(s). Methimazole 31-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 223-232 11027970-4 2000 Similarly, both inhibitors and methimazole did produce a significant decrease in 2-AF and m-PDA mutagenesis, when the activation system was S117, indicating that, similar to what occurs in mammalian systems, plant FMOs and cyt-P450s can metabolize aromatic amines to mutagenic product(s). aromatic amines 248-263 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 223-232 10997944-0 2000 Validation of bupropion hydroxylation as a selective marker of human cytochrome P450 2B6 catalytic activity. Bupropion 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-88 11012553-0 2000 In vitro evaluation of potential in vivo probes for human flavin-containing monooxygenase (FMO): metabolism of benzydamine and caffeine by FMO and P450 isoforms. Benzydamine 111-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-151 11012553-0 2000 In vitro evaluation of potential in vivo probes for human flavin-containing monooxygenase (FMO): metabolism of benzydamine and caffeine by FMO and P450 isoforms. Caffeine 127-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-151 11012553-1 2000 UNLABELLED: AIMS To determine the FMO and P450 isoform selectivity for metabolism of benzydamine and caffeine, two potential in vivo probes for human FMO. Benzydamine 85-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 11012553-1 2000 UNLABELLED: AIMS To determine the FMO and P450 isoform selectivity for metabolism of benzydamine and caffeine, two potential in vivo probes for human FMO. Caffeine 101-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 10997936-0 2000 CYP2B6 mediates the in vitro hydroxylation of bupropion: potential drug interactions with other antidepressants. Bupropion 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 10997936-4 2000 In microsomes containing cDNA-expressed CYPs, hydroxybupropion formation was mediated only by CYP2B6 at 50 microM bupropion (K(m) 85 microM). hydroxybupropion 46-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 10997944-1 2000 The purpose of this study was to establish bupropion (BUP) hydroxylation as a selective in vitro marker of cytochrome P450 (CYP) 2B6 catalytic activity. Bupropion 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-132 10997936-4 2000 In microsomes containing cDNA-expressed CYPs, hydroxybupropion formation was mediated only by CYP2B6 at 50 microM bupropion (K(m) 85 microM). Bupropion 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 10997936-5 2000 A CYP2B6 inhibitory antibody produced more than 95% inhibition of bupropion hydroxylation in four human livers. Bupropion 66-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 2-8 10997944-1 2000 The purpose of this study was to establish bupropion (BUP) hydroxylation as a selective in vitro marker of cytochrome P450 (CYP) 2B6 catalytic activity. Bupropion 54-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-132 10997936-6 2000 Bupropion hydroxylation activity at 250 microM was highly correlated with S-mephenytoin N-demethylation activity (yielding nirvanol), another CYP2B6-mediated reaction, in a panel of 32 human livers (r = 0.94). Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 10997944-6 2000 Among a panel of cDNA-expressed P450 isoforms, CYP2B6 and CYP2E1 demonstrated the highest rates of BUP hydroxylation at 12 mM BUP (7.0 and 2.4 pmol/min/pmol of P450, respectively). Bupropion 99-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 10997936-6 2000 Bupropion hydroxylation activity at 250 microM was highly correlated with S-mephenytoin N-demethylation activity (yielding nirvanol), another CYP2B6-mediated reaction, in a panel of 32 human livers (r = 0.94). Mephenytoin 74-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 10997944-6 2000 Among a panel of cDNA-expressed P450 isoforms, CYP2B6 and CYP2E1 demonstrated the highest rates of BUP hydroxylation at 12 mM BUP (7.0 and 2.4 pmol/min/pmol of P450, respectively). Bupropion 99-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 10997936-6 2000 Bupropion hydroxylation activity at 250 microM was highly correlated with S-mephenytoin N-demethylation activity (yielding nirvanol), another CYP2B6-mediated reaction, in a panel of 32 human livers (r = 0.94). ethylphenylhydantoin 123-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 10997944-6 2000 Among a panel of cDNA-expressed P450 isoforms, CYP2B6 and CYP2E1 demonstrated the highest rates of BUP hydroxylation at 12 mM BUP (7.0 and 2.4 pmol/min/pmol of P450, respectively). Bupropion 99-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 160-164 10997936-7 2000 The CYP2B6 content of 12 human livers highly correlated with bupropion hydroxylation activity (r = 0.96). Bupropion 61-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 10950858-1 2000 Serum of rabbits with a turpentine-induced acute inflammatory reaction (RS(INFLA)) and serum of humans with a viral infection (HS(INF)) were previously shown to diminish hepatic cytochrome P450 (P450) content and activity. Turpentine 24-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-193 10997936-8 2000 Thus bupropion hydroxylation is mediated almost exclusively by CYP2B6 and can serve as an index reaction reflecting activity of this isoform. Bupropion 5-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 11067776-10 2000 CYP2E1 was strongly correlated with chloral hydrate formation from trichloroethylene; CYP2B displayed the strongest correlation with trichloroethanol formation. 2,2,2-trichloroethanol 133-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-91 10950858-0 2000 Role of reactive oxygen intermediates in the decrease of hepatic cytochrome P450 activity by serum of humans and rabbits with an acute inflammatory reaction. Oxygen 17-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 10950858-1 2000 Serum of rabbits with a turpentine-induced acute inflammatory reaction (RS(INFLA)) and serum of humans with a viral infection (HS(INF)) were previously shown to diminish hepatic cytochrome P450 (P450) content and activity. Turpentine 24-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-199 10950858-4 2000 Exposure of H(CONT) or H(INFLA) to hydrogen peroxide (0.01-1.0 mM) and sodium nitroprusside (0.01-1.0 mM) produced a dose-dependent decrease in P450 content and in the formation of theophylline metabolites without modifying the amount of CYP1A1 and CYP1A2, whereas lipid peroxidation increased. Hydrogen Peroxide 35-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-148 10950858-4 2000 Exposure of H(CONT) or H(INFLA) to hydrogen peroxide (0.01-1.0 mM) and sodium nitroprusside (0.01-1.0 mM) produced a dose-dependent decrease in P450 content and in the formation of theophylline metabolites without modifying the amount of CYP1A1 and CYP1A2, whereas lipid peroxidation increased. Nitroprusside 71-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-148 10950858-4 2000 Exposure of H(CONT) or H(INFLA) to hydrogen peroxide (0.01-1.0 mM) and sodium nitroprusside (0.01-1.0 mM) produced a dose-dependent decrease in P450 content and in the formation of theophylline metabolites without modifying the amount of CYP1A1 and CYP1A2, whereas lipid peroxidation increased. Theophylline 181-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-148 10950858-7 2000 It is concluded that reactive oxygen intermediates are implicated in the decrease of H(INFLA) P450 content and activity induced by 4 h of exposure to RS(INFLA) or HS(INF). Oxygen 30-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-98 10945868-0 2000 Recombinant cytochrome P450 2D18 metabolism of dopamine and arachidonic acid. Dopamine 47-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 10945868-0 2000 Recombinant cytochrome P450 2D18 metabolism of dopamine and arachidonic acid. Arachidonic Acid 60-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 10945868-7 2000 Because the four aforementioned pharmaceutical substrates work by binding to neurotransmitter receptors, binding assays and oxidation reactions were performed to test whether dopamine is a substrate for P450 2D18. Dopamine 175-183 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 203-207 10945868-8 2000 These data indicate a K(S) value of 678 microM and that P450 2D18 can support the oxidation of dopamine to aminochrome through a peroxide-shunt mechanism. Dopamine 95-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 10945868-8 2000 These data indicate a K(S) value of 678 microM and that P450 2D18 can support the oxidation of dopamine to aminochrome through a peroxide-shunt mechanism. Peroxides 129-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 10945868-9 2000 We also report the P450 2D18-mediated omega-hydroxylation and epoxygenation of arachidonic acid, primarily leading to the formation of 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids, compounds that have been shown to have vasoactive properties in brain, kidney, and heart tissues. Arachidonic Acid 79-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 10945868-9 2000 We also report the P450 2D18-mediated omega-hydroxylation and epoxygenation of arachidonic acid, primarily leading to the formation of 8,9-, 11,12-, and 14,15-epoxyeicosatrienoic acids, compounds that have been shown to have vasoactive properties in brain, kidney, and heart tissues. -epoxyeicosatrienoic acids 158-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 10945868-10 2000 The data presented herein suggest a possible role for P450 involvement in membrane and receptor regulation via epoxyeicosatrienoic acid formation and a potential involvement of P450 in the oxidation of dopamine to reactive oxygen species under aberrant physiological conditions where the sequestering of dopamine becomes compromised, such as in Parkinson"s disease. epoxyeicosatrienoic acid 111-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 10945868-10 2000 The data presented herein suggest a possible role for P450 involvement in membrane and receptor regulation via epoxyeicosatrienoic acid formation and a potential involvement of P450 in the oxidation of dopamine to reactive oxygen species under aberrant physiological conditions where the sequestering of dopamine becomes compromised, such as in Parkinson"s disease. Dopamine 202-210 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 10945868-10 2000 The data presented herein suggest a possible role for P450 involvement in membrane and receptor regulation via epoxyeicosatrienoic acid formation and a potential involvement of P450 in the oxidation of dopamine to reactive oxygen species under aberrant physiological conditions where the sequestering of dopamine becomes compromised, such as in Parkinson"s disease. Dopamine 202-210 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-181 10945868-10 2000 The data presented herein suggest a possible role for P450 involvement in membrane and receptor regulation via epoxyeicosatrienoic acid formation and a potential involvement of P450 in the oxidation of dopamine to reactive oxygen species under aberrant physiological conditions where the sequestering of dopamine becomes compromised, such as in Parkinson"s disease. Reactive Oxygen Species 214-237 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 10945868-10 2000 The data presented herein suggest a possible role for P450 involvement in membrane and receptor regulation via epoxyeicosatrienoic acid formation and a potential involvement of P450 in the oxidation of dopamine to reactive oxygen species under aberrant physiological conditions where the sequestering of dopamine becomes compromised, such as in Parkinson"s disease. Reactive Oxygen Species 214-237 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-181 10945868-10 2000 The data presented herein suggest a possible role for P450 involvement in membrane and receptor regulation via epoxyeicosatrienoic acid formation and a potential involvement of P450 in the oxidation of dopamine to reactive oxygen species under aberrant physiological conditions where the sequestering of dopamine becomes compromised, such as in Parkinson"s disease. Dopamine 304-312 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 10859153-11 2000 The formation of 3-F-4-OHCIS was mainly catalyzed by CYP2C8 (V = 0.71 +/- 0.24 pmol/min/pmol of P450) and that of 4-F-2-OHCIS by CYP3A4 (0.16 +/- 0.03 pmol/min/pmol of P450). 3-f-4-ohcis 17-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-100 11051567-10 2000 The structure of cytochrome P450cam (P450) co-crystallized with a prototypal sensitizer-substrate, [Ru-C9-Ad]Cl2, has been determined (Dmochowski et al., Proc. [ru-c9-ad]cl2 99-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-41 10924340-4 2000 In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. Rifampin 33-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 10924340-4 2000 In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. Phenobarbital 49-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 10918309-5 2000 Larvae from polyphenol-rich habitats had a higher tolerance for polyphenols and higher midgut cytochrome P450 and esterase activities than larvae from polyphenol-poor habitats. Polyphenols 12-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-122 10859153-11 2000 The formation of 3-F-4-OHCIS was mainly catalyzed by CYP2C8 (V = 0.71 +/- 0.24 pmol/min/pmol of P450) and that of 4-F-2-OHCIS by CYP3A4 (0.16 +/- 0.03 pmol/min/pmol of P450). 3-f-4-ohcis 17-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 168-172 10859153-11 2000 The formation of 3-F-4-OHCIS was mainly catalyzed by CYP2C8 (V = 0.71 +/- 0.24 pmol/min/pmol of P450) and that of 4-F-2-OHCIS by CYP3A4 (0.16 +/- 0.03 pmol/min/pmol of P450). 4-f-2-ohcis 114-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-100 10867234-3 2000 GenBank comparison (BLAST) showed that the protein was closely related to P450 because a heme-binding region, which is highly conserved in all P450 sequences, was found in the ORF protein. Heme 89-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-78 10867234-3 2000 GenBank comparison (BLAST) showed that the protein was closely related to P450 because a heme-binding region, which is highly conserved in all P450 sequences, was found in the ORF protein. Heme 89-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 143-147 10860550-5 2000 This P450 exhibited omega-hydroxylation activity toward laurate, with a turnover number of 14.7 nmol/min/nmol of P450. Laurates 56-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 10799646-13 2000 CYP2B1 and its human homologue CYP2B6 also activate tobacco smoke procarcinogens such as NNK [4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone]. 4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone 94-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 10799653-8 2000 The most important isozyme for this reaction appeared to be CYP2B6, the microsomal content of which was found to be strongly correlated to N-deethylation of MDE (r(s) = 0.90; P < 0.001). Nitrogen 139-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 10860550-5 2000 This P450 exhibited omega-hydroxylation activity toward laurate, with a turnover number of 14.7 nmol/min/nmol of P450. Laurates 56-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-117 10860550-7 2000 It also showed omega-hydroxylation activity toward palmitate, with a turnover number of 0.78 nmol/min/nmol of P450. Palmitates 51-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-114 10788445-4 2000 This motif resembles phenobarbital response elements in the flanking regions of three phenobarbital-inducible genes, rat CYP2B2, mouse Cyp2b10, and human CYP2B6. Phenobarbital 21-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-160 10820139-8 2000 The mean activities of microsomal CYP1A2, CYP2B6, and CYP3A4/5 from modafinil-treated hepatocytes were higher (up to 2-fold) than those in the solvent-treated controls but were less than those produced by reference inducers of these enzymes. Modafinil 68-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 10820139-10 2000 Overall, modafinil was shown to have effects on human hepatic CYP1A2, CYP2B6, CYP2C9, CYP2C19, and CYP3A4/5 activities in vitro. Modafinil 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 11948012-1 2002 In vertebrates the wide variety of cytochromes P(450) (P(450)) is a key for elimination of low molecular weight xenobiotics and for the production and metabolism of steroid hormones. Steroids 165-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-61 10788445-4 2000 This motif resembles phenobarbital response elements in the flanking regions of three phenobarbital-inducible genes, rat CYP2B2, mouse Cyp2b10, and human CYP2B6. Phenobarbital 86-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-160 10821163-3 2000 The results indicate that sulphaphenazole, quinidine and alpha-naphthoflavone are selective inhibitors of CYP2C9 (IC50 = 0.5-0.7 microM), CYP2D6 (0.3-0.4 microM) and CYP1A (0.05-5 microM) respectively on the basis of the IC50, which are much lower than those of other P450 isoforms (> 10-fold). Sulfaphenazole 26-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 268-272 10692561-0 2000 Role of human liver microsomal CYP3A4 and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfamide. Nitrogen 63-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 10692561-0 2000 Role of human liver microsomal CYP3A4 and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfamide. Cyclophosphamide 87-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 10692561-0 2000 Role of human liver microsomal CYP3A4 and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfamide. Ifosfamide 108-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 10692561-6 2000 Because CYP2B6 can make a significant contribution to human liver microsomal IFA N-dechloroethylation, but only a minor contribution to IFA 4-hydroxylation, the selective inhibition of hepatic CYP2B6 activity in individuals with a high hepatic CYP2B6 content may provide a useful approach to minimize the formation of therapeutically inactive but toxic N-dechloroethylated IFA metabolites. Nitrogen 81-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 10729196-12 2000 Upon incubation with phenobarbital and rifampin (rifampicin), human hepatocytes increased CYP 2B6, 3A4, and 3A5 among others. Phenobarbital 21-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-97 10729196-12 2000 Upon incubation with phenobarbital and rifampin (rifampicin), human hepatocytes increased CYP 2B6, 3A4, and 3A5 among others. Rifampin 39-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-97 10729196-12 2000 Upon incubation with phenobarbital and rifampin (rifampicin), human hepatocytes increased CYP 2B6, 3A4, and 3A5 among others. Rifampin 49-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-97 10806389-7 2000 In this respect, it resembles the P450 isolated from acidothermophilic archaebacteria [McLean, M.A., Maves, S.A., Weiss, K.E., Krepich, S. & Sligar, S.G. (1998) Biochem. Adenosine Monophosphate 140-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 10784435-3 2000 CYP2C19 and CYP2B6 catalyzed most efficiently the aminopyrine N-demethylation, followed by CYP2C8 and CYP2D6. Aminopyrine 50-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 10784435-4 2000 Bisphenol A (1 mM) most efficiently inhibited aminopyrine N-demethylation by CYP2C8 and CYP2C19 by 82% and 85%, respectively, whereas inhibition of the activities by CYP 2B6 and 2D6 was less than 40%. bisphenol A 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-173 10821163-3 2000 The results indicate that sulphaphenazole, quinidine and alpha-naphthoflavone are selective inhibitors of CYP2C9 (IC50 = 0.5-0.7 microM), CYP2D6 (0.3-0.4 microM) and CYP1A (0.05-5 microM) respectively on the basis of the IC50, which are much lower than those of other P450 isoforms (> 10-fold). Quinidine 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 268-272 10821163-3 2000 The results indicate that sulphaphenazole, quinidine and alpha-naphthoflavone are selective inhibitors of CYP2C9 (IC50 = 0.5-0.7 microM), CYP2D6 (0.3-0.4 microM) and CYP1A (0.05-5 microM) respectively on the basis of the IC50, which are much lower than those of other P450 isoforms (> 10-fold). alpha-naphthoflavone 57-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 268-272 10821163-8 2000 Diethyldithiocarbamate, orphenadrine and furafylline were shown separately to be less selective inhibitors of CYP2E1, CYP2B6 and CYP1A isoforms by a broad range of IC50 that overlap those observed with other P450 isoforms. Ditiocarb 0-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 10821163-8 2000 Diethyldithiocarbamate, orphenadrine and furafylline were shown separately to be less selective inhibitors of CYP2E1, CYP2B6 and CYP1A isoforms by a broad range of IC50 that overlap those observed with other P450 isoforms. Ditiocarb 0-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 208-212 10821163-8 2000 Diethyldithiocarbamate, orphenadrine and furafylline were shown separately to be less selective inhibitors of CYP2E1, CYP2B6 and CYP1A isoforms by a broad range of IC50 that overlap those observed with other P450 isoforms. Orphenadrine 24-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 10821163-8 2000 Diethyldithiocarbamate, orphenadrine and furafylline were shown separately to be less selective inhibitors of CYP2E1, CYP2B6 and CYP1A isoforms by a broad range of IC50 that overlap those observed with other P450 isoforms. Orphenadrine 24-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 208-212 10821163-8 2000 Diethyldithiocarbamate, orphenadrine and furafylline were shown separately to be less selective inhibitors of CYP2E1, CYP2B6 and CYP1A isoforms by a broad range of IC50 that overlap those observed with other P450 isoforms. furafylline 41-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 10821163-8 2000 Diethyldithiocarbamate, orphenadrine and furafylline were shown separately to be less selective inhibitors of CYP2E1, CYP2B6 and CYP1A isoforms by a broad range of IC50 that overlap those observed with other P450 isoforms. furafylline 41-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 208-212 10752639-0 2000 In vitro inhibition of human P450 enzymes by prenylated flavonoids from hops, Humulus lupulus. Flavonoids 56-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 11249525-5 2000 Differences in the metabolic balance between hepatic P450 (especially CYP 1A2) and MAO-A inactivation lead to potential drug interactions for all TELs with the oral contraceptive pill (OCP), fluvoxamine and the quinilone antibiotics (with increased triptan levels). quinilone 211-220 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 11249525-5 2000 Differences in the metabolic balance between hepatic P450 (especially CYP 1A2) and MAO-A inactivation lead to potential drug interactions for all TELs with the oral contraceptive pill (OCP), fluvoxamine and the quinilone antibiotics (with increased triptan levels). Tryptamines 249-256 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 10718780-5 2000 Desethylamiodarone competitively inhibited the catalytic activities of CYP2D6 (Ki=4.5 microm ) and noncompetitively inhibited CYP2A6 (Ki=13.5 microm ), CYP2B6 (Ki=5.4 microm ), and CYP3A4 (Ki=12.1 microm ). desethylamiodarone 0-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-158 10718780-8 2000 Amiodarone inactivated CYP3A4, while desethylamiodarone inactivated CYP1A1, CYP1A2, CYP2B6, and CYP2D6. desethylamiodarone 37-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 10752642-6 2000 Primary cultures of human hepatocytes were used to investigate the induction potential of troglitazone with respect to CYP3A4, CYP2B6 and CYP1A1/2. Troglitazone 90-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 10752642-9 2000 Troglitazone increased CYP2B6 immunoreactive protein but did not significantly effect CYP1A1/2 activity, immunoreactive protein or mRNA. Troglitazone 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-29 10752639-4 2000 Additionally, preliminary studies have shown these flavonoids (at 100 microM) to be inhibitory of P450-mediated activation reactions in a variety of in vitro systems. Flavonoids 51-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 10752642-13 2000 These results provide evidence that troglitazone can induce CYP3A and CYP2B enzymes while apparently not altering CYP1A. Troglitazone 36-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-75 10752639-19 2000 These results suggest that the hop flavonoids are potent and selective inhibitors of human cytochrome P450 and warrant further in vivo investigations. Flavonoids 35-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-106 10640505-4 2000 The results show that estrone and 17beta-estadiol were converted into the corresponding quinols by CYP1A1, CYP2B6, and CYP2E1. Estrone 22-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 10698112-9 2000 In contrast to iNOS we found, however, that in P450 partially negative propionate side chains of protoporphyrin IX are located on the opposite side of the heme plane. protoporphyrin IX 97-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 10698112-9 2000 In contrast to iNOS we found, however, that in P450 partially negative propionate side chains of protoporphyrin IX are located on the opposite side of the heme plane. Heme 155-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 10698112-10 2000 As a result of this and the absence of other negatively charged residues the distal (substrate binding) side of P450 should be less negative than that of NOS and therefore its affinity toward the partially positive arginine is reduced. Arginine 215-223 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 10805063-8 2000 Barnidipine and amlodipine inhibited the CYP2B6 activity. mepirodipine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 10805063-8 2000 Barnidipine and amlodipine inhibited the CYP2B6 activity. Amlodipine 16-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 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 2 subfamily B member 6 Homo sapiens 138-144 10888271-0 2000 Interaction of histamine and other bioamines with cytochromes P450: implications for cell growth modulation and chemopotentiation by drugs. Histamine 15-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 10888271-0 2000 Interaction of histamine and other bioamines with cytochromes P450: implications for cell growth modulation and chemopotentiation by drugs. bioamines 35-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 10888271-4 2000 We have shown that polyamines, hormones (including estrogen, testosterone and progesterone), antihormones (including tamoxifen and flutamide) and various antidepressants and antihistamines, all inhibit histamine binding to P450; we have postulated that, through binding to the heme moiety, intracellular histamine regulates cell function by modulating the catalytic activity of P450 enzymes, an action that may be perturbed by endogenous and exogenous substances. Polyamines 19-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 223-227 10888271-4 2000 We have shown that polyamines, hormones (including estrogen, testosterone and progesterone), antihormones (including tamoxifen and flutamide) and various antidepressants and antihistamines, all inhibit histamine binding to P450; we have postulated that, through binding to the heme moiety, intracellular histamine regulates cell function by modulating the catalytic activity of P450 enzymes, an action that may be perturbed by endogenous and exogenous substances. Polyamines 19-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 378-382 10888271-4 2000 We have shown that polyamines, hormones (including estrogen, testosterone and progesterone), antihormones (including tamoxifen and flutamide) and various antidepressants and antihistamines, all inhibit histamine binding to P450; we have postulated that, through binding to the heme moiety, intracellular histamine regulates cell function by modulating the catalytic activity of P450 enzymes, an action that may be perturbed by endogenous and exogenous substances. Progesterone 78-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 223-227 10888271-4 2000 We have shown that polyamines, hormones (including estrogen, testosterone and progesterone), antihormones (including tamoxifen and flutamide) and various antidepressants and antihistamines, all inhibit histamine binding to P450; we have postulated that, through binding to the heme moiety, intracellular histamine regulates cell function by modulating the catalytic activity of P450 enzymes, an action that may be perturbed by endogenous and exogenous substances. Tamoxifen 117-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 223-227 10888271-4 2000 We have shown that polyamines, hormones (including estrogen, testosterone and progesterone), antihormones (including tamoxifen and flutamide) and various antidepressants and antihistamines, all inhibit histamine binding to P450; we have postulated that, through binding to the heme moiety, intracellular histamine regulates cell function by modulating the catalytic activity of P450 enzymes, an action that may be perturbed by endogenous and exogenous substances. Flutamide 131-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 223-227 10888271-4 2000 We have shown that polyamines, hormones (including estrogen, testosterone and progesterone), antihormones (including tamoxifen and flutamide) and various antidepressants and antihistamines, all inhibit histamine binding to P450; we have postulated that, through binding to the heme moiety, intracellular histamine regulates cell function by modulating the catalytic activity of P450 enzymes, an action that may be perturbed by endogenous and exogenous substances. Flutamide 131-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 378-382 10888271-4 2000 We have shown that polyamines, hormones (including estrogen, testosterone and progesterone), antihormones (including tamoxifen and flutamide) and various antidepressants and antihistamines, all inhibit histamine binding to P450; we have postulated that, through binding to the heme moiety, intracellular histamine regulates cell function by modulating the catalytic activity of P450 enzymes, an action that may be perturbed by endogenous and exogenous substances. Heme 277-281 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 223-227 10888271-4 2000 We have shown that polyamines, hormones (including estrogen, testosterone and progesterone), antihormones (including tamoxifen and flutamide) and various antidepressants and antihistamines, all inhibit histamine binding to P450; we have postulated that, through binding to the heme moiety, intracellular histamine regulates cell function by modulating the catalytic activity of P450 enzymes, an action that may be perturbed by endogenous and exogenous substances. Histamine 202-211 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 223-227 10888271-4 2000 We have shown that polyamines, hormones (including estrogen, testosterone and progesterone), antihormones (including tamoxifen and flutamide) and various antidepressants and antihistamines, all inhibit histamine binding to P450; we have postulated that, through binding to the heme moiety, intracellular histamine regulates cell function by modulating the catalytic activity of P450 enzymes, an action that may be perturbed by endogenous and exogenous substances. Histamine 202-211 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 378-382 10888271-5 2000 We now demonstrate that, in addition to histamine, melatonin and the biogenic amines dopamine, serotonin and noradrenaline bind to P450 isozymes and to cytochrome C. Melatonin 51-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-135 10888271-5 2000 We now demonstrate that, in addition to histamine, melatonin and the biogenic amines dopamine, serotonin and noradrenaline bind to P450 isozymes and to cytochrome C. Dopamine 85-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-135 10888271-5 2000 We now demonstrate that, in addition to histamine, melatonin and the biogenic amines dopamine, serotonin and noradrenaline bind to P450 isozymes and to cytochrome C. Serotonin 95-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-135 10888271-5 2000 We now demonstrate that, in addition to histamine, melatonin and the biogenic amines dopamine, serotonin and noradrenaline bind to P450 isozymes and to cytochrome C. Norepinephrine 109-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-135 10640505-4 2000 The results show that estrone and 17beta-estadiol were converted into the corresponding quinols by CYP1A1, CYP2B6, and CYP2E1. 17beta-estadiol 34-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 10640505-4 2000 The results show that estrone and 17beta-estadiol were converted into the corresponding quinols by CYP1A1, CYP2B6, and CYP2E1. Hydroquinones 88-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 10570024-3 1999 The apparent K(m) values of human liver microsomes for S-mephobarbital N-demethylation were close to that of cDNA-expressed CYP2B6 (about 250 microM). Mephobarbital 55-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-130 10755318-9 2000 RESULTS: (1) DPPE evoked "type I" (substrate site binding) absorbance-difference spectra with CYP2D6 (K(S) = 4.1 +/- 0.4 microM), CYP3A4 (K(S) = 31 +/- 15 microM) and CYP1A1 (K(S) = 40 +/- 9 microM), but not with CYP2B6. tesmilifene 13-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 213-219 11844358-5 2000 Molecular genetic studies have demonstrated that CYP11a, the gene encoding P450 cholesterol side chain cleavage, is a major susceptibility locus for development of hyperandrogenism in PCOS. Cholesterol 80-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-79 10659951-2 2000 Investigated were the effects of a new oral antidiabetic drug, troglitazone, and its three metabolites and antidiabetic drug candidates pioglitazone and rosiglitazone on xenobiotic oxidations catalyzed by nine recombinant human cytochrome P450 (P450 or CYP) enzymes and by human liver microsomes. Troglitazone 63-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 228-256 10570024-3 1999 The apparent K(m) values of human liver microsomes for S-mephobarbital N-demethylation were close to that of cDNA-expressed CYP2B6 (about 250 microM). Nitrogen 71-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-130 10570024-4 1999 The N-demethylase activity of S-mephobarbital in 10 human liver microsomes was strongly correlated with immunodetectable CYP2B6 levels (r = 0.920, p<.001). Mephobarbital 30-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-127 10570024-5 1999 Orphenadrine (300 microM), a CYP2B6 inhibitor, inhibited the N-demethylase activity of S-mephobarbital in human liver microsomes to 29% of control activity. Orphenadrine 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 10570024-5 1999 Orphenadrine (300 microM), a CYP2B6 inhibitor, inhibited the N-demethylase activity of S-mephobarbital in human liver microsomes to 29% of control activity. Mephobarbital 87-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 10570024-6 1999 Therefore, it appears that CYP2B6 mainly catalyzes S-mephobarbital N-demethylation in human liver microsomes. Mephobarbital 51-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 10570024-6 1999 Therefore, it appears that CYP2B6 mainly catalyzes S-mephobarbital N-demethylation in human liver microsomes. Nitrogen 67-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 10630892-6 1999 Human lymphoblast cell microsomes expressing human CYP2B6 incubated with the steroids investigated produced traces of 6beta-hydroxyl metabolites with testosterone and 17alpha-methyltestosterone only. Steroids 77-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 10647906-5 1999 Regarding ethoxyresorufin O-deethylation catalysed by CYP1A1 and benzyloxyresorufin O-dealkylation by CYP2B6, the subtle change of a substitution of the 1,4-benzothiazine structure affected the inhibition selectivity. ethoxyresorufin 10-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 10647906-5 1999 Regarding ethoxyresorufin O-deethylation catalysed by CYP1A1 and benzyloxyresorufin O-dealkylation by CYP2B6, the subtle change of a substitution of the 1,4-benzothiazine structure affected the inhibition selectivity. benzyloxyresorufin 65-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 10647906-5 1999 Regarding ethoxyresorufin O-deethylation catalysed by CYP1A1 and benzyloxyresorufin O-dealkylation by CYP2B6, the subtle change of a substitution of the 1,4-benzothiazine structure affected the inhibition selectivity. 1,4-benzothiazine 153-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 10570242-6 1999 Unlike the microsomal P-450 reductase, the FMN semiquinone is the active electron donor to the heme iron in P-450(BM-3). flavin mononucleotide semiquinone 43-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-118 10570242-6 1999 Unlike the microsomal P-450 reductase, the FMN semiquinone is the active electron donor to the heme iron in P-450(BM-3). Heme 95-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-118 10570242-6 1999 Unlike the microsomal P-450 reductase, the FMN semiquinone is the active electron donor to the heme iron in P-450(BM-3). Iron 100-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-118 10570242-7 1999 The crystal structure of P-450(BM-3) heme/FMN bidomain provides important insights into why the FMN semiquinone is the preferred electron donor to the heme as well as how substrate-induced structural changes possibly affect the FMN and heme domain-domain interaction. semiquinone 100-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-35 10570242-7 1999 The crystal structure of P-450(BM-3) heme/FMN bidomain provides important insights into why the FMN semiquinone is the preferred electron donor to the heme as well as how substrate-induced structural changes possibly affect the FMN and heme domain-domain interaction. Heme 37-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-35 10570242-7 1999 The crystal structure of P-450(BM-3) heme/FMN bidomain provides important insights into why the FMN semiquinone is the preferred electron donor to the heme as well as how substrate-induced structural changes possibly affect the FMN and heme domain-domain interaction. Flavin Mononucleotide 42-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-35 10630892-6 1999 Human lymphoblast cell microsomes expressing human CYP2B6 incubated with the steroids investigated produced traces of 6beta-hydroxyl metabolites with testosterone and 17alpha-methyltestosterone only. 6beta-hydroxyl 118-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 10570242-7 1999 The crystal structure of P-450(BM-3) heme/FMN bidomain provides important insights into why the FMN semiquinone is the preferred electron donor to the heme as well as how substrate-induced structural changes possibly affect the FMN and heme domain-domain interaction. Heme 151-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-35 10558891-0 1999 Formation of indigo by recombinant mammalian cytochrome P450. Indigo Carmine 13-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 10558891-6 1999 Accordingly, we speculated that indole, formed as a breakdown product of tryptophan in bacteria, was hydroxylated by the P450 system, leading to indigo formation. indole 32-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 10630892-6 1999 Human lymphoblast cell microsomes expressing human CYP2B6 incubated with the steroids investigated produced traces of 6beta-hydroxyl metabolites with testosterone and 17alpha-methyltestosterone only. Testosterone 150-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 10558891-6 1999 Accordingly, we speculated that indole, formed as a breakdown product of tryptophan in bacteria, was hydroxylated by the P450 system, leading to indigo formation. Tryptophan 73-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 10558891-6 1999 Accordingly, we speculated that indole, formed as a breakdown product of tryptophan in bacteria, was hydroxylated by the P450 system, leading to indigo formation. Indigo Carmine 145-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 10630892-6 1999 Human lymphoblast cell microsomes expressing human CYP2B6 incubated with the steroids investigated produced traces of 6beta-hydroxyl metabolites with testosterone and 17alpha-methyltestosterone only. Methyltestosterone 167-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 10558891-7 1999 Bacterial membranes containing recombinant P450 2E1 and human NPR were incubated in vitro with indole and shown to catalyze formation of a blue pigment in a time- and cofactor-dependent manner. indole 95-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 10760842-6 1999 Only CYP2B6, 2C8 and 2C18 were considered likely candidates as contributors to residual 3-methoxymorphinan activity. 3-methoxymorphinan 88-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-11 10558891-8 1999 These studies suggest potential applications of mammalian P450 enzymes in industrial indigo production or in the development of novel colorimetric assays based on indole hydroxylation. Indigo Carmine 85-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 10558891-8 1999 These studies suggest potential applications of mammalian P450 enzymes in industrial indigo production or in the development of novel colorimetric assays based on indole hydroxylation. indole 163-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 10714368-6 1999 This effect is the result of an impairment involving FSH stimulation of G protein-coupled receptors (GPCR) and cyclic AMP-mediated activation of aromatase cytochrome P450 gene expression. Cyclic AMP 111-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-170 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. artemisinin 95-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-128 10583023-5 1999 RESULTS: The involvement of CYP2B6 in artemisinin metabolism was demonstrated by metabolism of artemisinin by recombinant CYP2B6, inhibition of artemisinin disappearance in human liver microsomes by orphenadrine (76%) and primary inclusion of CYP2B6 in the tree-based regression model. Orphenadrine 199-211 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 10557259-3 1999 We have found that Ru photosensitizers linked to P450 substrates specifically recognize submicromolar cytochrome P450(cam) in the presence of other heme proteins. cafestol palmitate 118-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 10557259-3 1999 We have found that Ru photosensitizers linked to P450 substrates specifically recognize submicromolar cytochrome P450(cam) in the presence of other heme proteins. cafestol palmitate 118-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-117 10557259-3 1999 We have found that Ru photosensitizers linked to P450 substrates specifically recognize submicromolar cytochrome P450(cam) in the presence of other heme proteins. Heme 148-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 10557259-4 1999 In the P450:Ru-substrate conjugates, energy transfer to the heme dramatically accelerates the Ru-luminescence decay. Ruthenium 12-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 7-11 10557259-4 1999 In the P450:Ru-substrate conjugates, energy transfer to the heme dramatically accelerates the Ru-luminescence decay. Heme 60-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 7-11 10557259-5 1999 The crystal structure of a P450(cam):Ru-adamantyl complex reveals access to the active center via a channel whose depth (Ru-Fe distance is 21 A) is virtually the same as that extracted from an analysis of the energy-transfer kinetics. ru-adamantyl 37-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 10557259-5 1999 The crystal structure of a P450(cam):Ru-adamantyl complex reveals access to the active center via a channel whose depth (Ru-Fe distance is 21 A) is virtually the same as that extracted from an analysis of the energy-transfer kinetics. Ruthenium 37-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 10557259-5 1999 The crystal structure of a P450(cam):Ru-adamantyl complex reveals access to the active center via a channel whose depth (Ru-Fe distance is 21 A) is virtually the same as that extracted from an analysis of the energy-transfer kinetics. Iron 124-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 10534317-9 1999 CYPs 3A4 and 3A5 preferentially produced (R)N2-DCl-IFA and (R)N3-DCl-IFA (derived from R-IFA and S-IFA, respectively), whereas CYP2B6 correspondingly formed (S)N3-DCl-IFA and (S)N2-DCl-IFA. (s)n3-dcl-ifa 157-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 10534317-9 1999 CYPs 3A4 and 3A5 preferentially produced (R)N2-DCl-IFA and (R)N3-DCl-IFA (derived from R-IFA and S-IFA, respectively), whereas CYP2B6 correspondingly formed (S)N3-DCl-IFA and (S)N2-DCl-IFA. (s)n2-dcl-ifa 175-188 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-133 10534317-10 1999 In human liver microsomes, the CYP3A-specific inhibitor troleandomycin suppressed (R)N2- and (R)N3-DCl-IFA formation by >/=80%, whereas (S)N2- and (S)N3-DCl-IFA formation were selectively inhibited (>/=85%) by a CYP2B6-specific monoclonal antibody. Troleandomycin 56-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 218-224 10534317-11 1999 The overall extent of IFA N-dechloroethylation varied with the CYP3A4 and CYP2B6 content of each liver, but was significantly lower for R-IFA (32 +/- 13%) than for S-IFA (62 +/- 17%, n = 8; p <.001) in all livers examined. Nitrogen 26-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 10512767-5 1999 Furthermore, this activity could be stimulated in the presence of either alpha-benzoflavone or beta-benzoflavone in an analogous manner to that reported for mammalian P450 forms including human liver cytochrome P4503A4 (CYP3A4). beta-benzoflavone 95-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 167-171 10583023-10 1999 The rate of artemisinin metabolism in recombinant CYP2A6 was 15% of that for recombinant CYP2B6. artemisinin 12-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-95 10583023-12 1999 CONCLUSIONS: Artemisinin metabolism in human liver microsomes is mediated primarily by CYP2B6 with probable secondary contribution of CYP3A4 in individuals with low CYP2B6 expression. artemisinin 13-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 10583023-12 1999 CONCLUSIONS: Artemisinin metabolism in human liver microsomes is mediated primarily by CYP2B6 with probable secondary contribution of CYP3A4 in individuals with low CYP2B6 expression. artemisinin 13-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-171 10760842-7 1999 The residual 3-methoxymorphinan activity was highly correlated with CYP2B6 activity as measured by CYP2B6 antibody (r(2)=0.90, p<0.001) and by orphenadrine (r(2)=0.97, p<0.001), but was not correlated (r(2)=0.12, p>0.05) with CYP2C8 activity. 3-methoxymorphinan 13-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 10760842-7 1999 The residual 3-methoxymorphinan activity was highly correlated with CYP2B6 activity as measured by CYP2B6 antibody (r(2)=0.90, p<0.001) and by orphenadrine (r(2)=0.97, p<0.001), but was not correlated (r(2)=0.12, p>0.05) with CYP2C8 activity. 3-methoxymorphinan 13-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 10760842-8 1999 Collectively, these findings suggest that CYP2B6 is a major contributor towards residual 3-methoxymorphinan activity, while CYP2C8 and 2C18 are either minor contributors or do not contribute to this metabolic process. 3-methoxymorphinan 89-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 10462436-4 1999 Two other nuclear receptors, designated LXR and FXR, which are respectively activated by oxysterols and bile acids, also play a role in liver P450 expression, in this case regulation of P450 cholesterol 7alpha-hydroxylase, a key enzyme of bile acid biosynthesis. Oxysterols 89-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 10449188-10 1999 Comparison of data obtained with CYP-expressing cells and human hepatocytes suggests that CYP2C8 > CYP2C19 approximately CYP2C18 >> CYP2B6 are the isoforms implicated in the 5-hydroxylation of diclofenac in vivo. Diclofenac 202-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 10462436-4 1999 Two other nuclear receptors, designated LXR and FXR, which are respectively activated by oxysterols and bile acids, also play a role in liver P450 expression, in this case regulation of P450 cholesterol 7alpha-hydroxylase, a key enzyme of bile acid biosynthesis. Oxysterols 89-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 186-190 10462436-4 1999 Two other nuclear receptors, designated LXR and FXR, which are respectively activated by oxysterols and bile acids, also play a role in liver P450 expression, in this case regulation of P450 cholesterol 7alpha-hydroxylase, a key enzyme of bile acid biosynthesis. Bile Acids and Salts 104-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 10462436-4 1999 Two other nuclear receptors, designated LXR and FXR, which are respectively activated by oxysterols and bile acids, also play a role in liver P450 expression, in this case regulation of P450 cholesterol 7alpha-hydroxylase, a key enzyme of bile acid biosynthesis. Bile Acids and Salts 104-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 186-190 10462436-4 1999 Two other nuclear receptors, designated LXR and FXR, which are respectively activated by oxysterols and bile acids, also play a role in liver P450 expression, in this case regulation of P450 cholesterol 7alpha-hydroxylase, a key enzyme of bile acid biosynthesis. Bile Acids and Salts 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-146 10462436-4 1999 Two other nuclear receptors, designated LXR and FXR, which are respectively activated by oxysterols and bile acids, also play a role in liver P450 expression, in this case regulation of P450 cholesterol 7alpha-hydroxylase, a key enzyme of bile acid biosynthesis. Bile Acids and Salts 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 186-190 10216279-4 1999 The Vmax for 6alpha-hydroxylation of taurochenodeoxycholic acid by CYP3A4 was 18.2 nmol/nmol P450/min and the apparent Km was 90 microM. 6alpha 13-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-101 10403825-5 1999 This P450 exhibited 6beta-hydroxylation activity toward both testosterone and progesterone. Testosterone 61-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 10403825-5 1999 This P450 exhibited 6beta-hydroxylation activity toward both testosterone and progesterone. Progesterone 78-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Phosphatidylcholines 135-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Phosphatidylcholines 135-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Phosphatidylcholines 135-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Phosphatidylcholines 135-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Phospholipids 169-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Phospholipids 169-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Phospholipids 169-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Phospholipids 169-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Protactinium 195-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Protactinium 195-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Protactinium 195-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Protactinium 195-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Choline 147-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Choline 147-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Choline 147-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10375402-2 1999 Purified human liver microsomal cytochromes P450 (P450)-P450 1A2 and P450 2E1-were shown to have appreciable PLD activity, hydrolyzing phosphatidylcholine but not other phospholipids, generating PA and choline. Choline 147-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 10383922-5 1999 Desmethylazelastine, but not azelastine and 6-hydroxyazelastine, uncompetitively inhibited CYP2B6 activity (Ki = 32.6 +/- 4.8 microM). desmethylazelastine 0-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 10383922-5 1999 Desmethylazelastine, but not azelastine and 6-hydroxyazelastine, uncompetitively inhibited CYP2B6 activity (Ki = 32.6 +/- 4.8 microM). azelastine 9-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 10471061-2 1999 Its expression in Saccharomyces cerevisiae enabled us to obtain, at a high level, an active yeast-expressed CYP2B6 protein, so as to assess its role in the metabolism of ethoxyresorufin, pentoxyresorufin, benzyloxyresorufin, ethoxycoumarin, testosterone and cyclophosphamide. ethoxyresorufin 170-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 10471061-2 1999 Its expression in Saccharomyces cerevisiae enabled us to obtain, at a high level, an active yeast-expressed CYP2B6 protein, so as to assess its role in the metabolism of ethoxyresorufin, pentoxyresorufin, benzyloxyresorufin, ethoxycoumarin, testosterone and cyclophosphamide. pentoxyresorufin 187-203 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 10471061-2 1999 Its expression in Saccharomyces cerevisiae enabled us to obtain, at a high level, an active yeast-expressed CYP2B6 protein, so as to assess its role in the metabolism of ethoxyresorufin, pentoxyresorufin, benzyloxyresorufin, ethoxycoumarin, testosterone and cyclophosphamide. benzyloxyresorufin 205-223 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 10471061-2 1999 Its expression in Saccharomyces cerevisiae enabled us to obtain, at a high level, an active yeast-expressed CYP2B6 protein, so as to assess its role in the metabolism of ethoxyresorufin, pentoxyresorufin, benzyloxyresorufin, ethoxycoumarin, testosterone and cyclophosphamide. 3-ethoxychromen-2-one 225-239 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 10471061-2 1999 Its expression in Saccharomyces cerevisiae enabled us to obtain, at a high level, an active yeast-expressed CYP2B6 protein, so as to assess its role in the metabolism of ethoxyresorufin, pentoxyresorufin, benzyloxyresorufin, ethoxycoumarin, testosterone and cyclophosphamide. Testosterone 241-253 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 10471061-2 1999 Its expression in Saccharomyces cerevisiae enabled us to obtain, at a high level, an active yeast-expressed CYP2B6 protein, so as to assess its role in the metabolism of ethoxyresorufin, pentoxyresorufin, benzyloxyresorufin, ethoxycoumarin, testosterone and cyclophosphamide. Cyclophosphamide 258-274 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 10517986-1 1999 To clarify the roles of human cytochrome P450 (P450 or CYP) 2A6 and 2E1 on the metabolic activation of N-nitrosamines, we established genetically engineered Salmonella typhimurium strains harboring human CYP2A6 or CYP2E1 together with NADPH-P450 reductase (OR). n-nitrosamines 103-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-71 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 2 subfamily B member 6 Homo sapiens 36-39 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 2 subfamily B member 6 Homo sapiens 144-147 10383917-6 1999 The anti-CYP2B6 antibody inhibited the sertraline N-demethylation activities by 35%. Sertraline 39-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 10383917-7 1999 Sertraline N-demethylation activities were detected in all cDNA-expressed CYP isoforms studied. Sertraline 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-77 10383917-8 1999 In particular, CYP2C19, CYP2B6, CYP2C9-Arg, CYP2D6-Val, and CYP3A4 all showed relatively high activity. Arginine 39-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 10383917-10 1999 The results suggest that at least five isoforms of CYP (CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A4) are involved in the sertraline N-demethylation in human liver microsomes and that the contribution of any individual isoform does not exceed 40% of overall metabolism. sertraline n 117-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-54 10383917-10 1999 The results suggest that at least five isoforms of CYP (CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A4) are involved in the sertraline N-demethylation in human liver microsomes and that the contribution of any individual isoform does not exceed 40% of overall metabolism. sertraline n 117-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 10348794-3 1999 CYP2B6 metabolized CPA at a approximately 16-fold higher in vitro intrinsic clearance (apparent Vmax/Km) than IFA, whereas 3A4 demonstrated approximately 2-fold higher Vmax/Km toward IFA. Cyclophosphamide 19-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 10348794-10 1999 These data further establish the significance of human liver CYP2B6 for the activation of the clinically important cancer chemotherapeutic prodrug CPA. Cyclophosphamide 147-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 10471061-3 1999 Kinetic analysis showed that human CYP2B6 preferentially metabolized benzyloxyresorufin and pentoxyresorufin, although other CYPs also metabolized these substrates in human liver microsomes. benzyloxyresorufin 69-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 10471061-3 1999 Kinetic analysis showed that human CYP2B6 preferentially metabolized benzyloxyresorufin and pentoxyresorufin, although other CYPs also metabolized these substrates in human liver microsomes. pentoxyresorufin 92-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 10471061-4 1999 CYP2B6 also manifested a strong 4-hydroxycyclophosphamide activity. 4-hydroxycyclophosphamide 32-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 10471061-11 1999 We also found that CYP2B6 is induced at protein and mRNA levels by phenobarbital (2 mM) and cyclophosphamide (1 mM), an anticancer drug known to be metabolized by CYP2B6. Phenobarbital 67-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 10471061-11 1999 We also found that CYP2B6 is induced at protein and mRNA levels by phenobarbital (2 mM) and cyclophosphamide (1 mM), an anticancer drug known to be metabolized by CYP2B6. Phenobarbital 67-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 10471061-11 1999 We also found that CYP2B6 is induced at protein and mRNA levels by phenobarbital (2 mM) and cyclophosphamide (1 mM), an anticancer drug known to be metabolized by CYP2B6. Cyclophosphamide 92-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 10471061-11 1999 We also found that CYP2B6 is induced at protein and mRNA levels by phenobarbital (2 mM) and cyclophosphamide (1 mM), an anticancer drug known to be metabolized by CYP2B6. Cyclophosphamide 92-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-169 10386659-3 1999 Three factor repeated measures multivariate analysis of variance was used to evaluate the effect of alcohol on the amplitudes and latencies of the P450 component to the happy and sad faces. Alcohols 100-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-151 10386659-4 1999 As compared to placebo, following alcohol ingestion, male subjects had decreased P450 amplitudes but only to male happy faces compared to female happy faces. Alcohols 34-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 10335450-3 1999 This strategy has been exemplified for the alkylating agents cyclophosphamide and ifosfamide, which are bioactivated by select P450 enzymes whose expression is generally high in liver and deficient in tumor cells. Cyclophosphamide 61-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 10335450-3 1999 This strategy has been exemplified for the alkylating agents cyclophosphamide and ifosfamide, which are bioactivated by select P450 enzymes whose expression is generally high in liver and deficient in tumor cells. Ifosfamide 82-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 10216279-4 1999 The Vmax for 6alpha-hydroxylation of taurochenodeoxycholic acid by CYP3A4 was 18.2 nmol/nmol P450/min and the apparent Km was 90 microM. Taurochenodeoxycholic Acid 37-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-101 10405640-9 1999 Taxanes are metabolized and inactivated by p450 isozymes, and this is related to drug-resistant to taxanes. Taxoids 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 10405640-9 1999 Taxanes are metabolized and inactivated by p450 isozymes, and this is related to drug-resistant to taxanes. Taxoids 99-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 10101147-10 1999 The antibody also was found to immunoinhibit CYP2B6-catalyzed N-demethylation of (S)-mephenytoin in human liver microsomes by 68 to 79%. Nitrogen 62-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 10197616-11 1999 These results, the first report on the comparative metabolism of mono-NP in humans, clearly demonstrate that the role of specific human P450 enzymes in catalyzing oxidative and reductive pathways of mono-NP is dependent upon the position of the nitro group. mono-np 65-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 10197616-11 1999 These results, the first report on the comparative metabolism of mono-NP in humans, clearly demonstrate that the role of specific human P450 enzymes in catalyzing oxidative and reductive pathways of mono-NP is dependent upon the position of the nitro group. mono-np 199-206 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 10197616-11 1999 These results, the first report on the comparative metabolism of mono-NP in humans, clearly demonstrate that the role of specific human P450 enzymes in catalyzing oxidative and reductive pathways of mono-NP is dependent upon the position of the nitro group. nitro 245-250 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 10197616-1 1999 Determining the capability of humans to metabolize the mononitropyrene (mono-NP) isomers 1-, 2-, and 4-NP and understanding which human cytochrome P450 (P450) enzymes are involved in their activation and/or detoxification is important in the assessment of individual susceptibility to these environmental carcinogens. mononitropyrene 55-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-151 10197616-1 1999 Determining the capability of humans to metabolize the mononitropyrene (mono-NP) isomers 1-, 2-, and 4-NP and understanding which human cytochrome P450 (P450) enzymes are involved in their activation and/or detoxification is important in the assessment of individual susceptibility to these environmental carcinogens. mononitropyrene 55-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 10197616-7 1999 The role of specific P450 enzymes in the human hepatic microsomal metabolism of mono-NP was investigated by correlating the P450-dependent catalytic activities in each microsomal sample with the levels of individual metabolites formed by the same microsomes and by examining the effects of agents that can either inhibit or stimulate specific P450 enzymes in mono-NP metabolism. mono-np 80-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 10197616-7 1999 The role of specific P450 enzymes in the human hepatic microsomal metabolism of mono-NP was investigated by correlating the P450-dependent catalytic activities in each microsomal sample with the levels of individual metabolites formed by the same microsomes and by examining the effects of agents that can either inhibit or stimulate specific P450 enzymes in mono-NP metabolism. mono-np 80-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 10197616-7 1999 The role of specific P450 enzymes in the human hepatic microsomal metabolism of mono-NP was investigated by correlating the P450-dependent catalytic activities in each microsomal sample with the levels of individual metabolites formed by the same microsomes and by examining the effects of agents that can either inhibit or stimulate specific P450 enzymes in mono-NP metabolism. mono-np 80-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 10197616-7 1999 The role of specific P450 enzymes in the human hepatic microsomal metabolism of mono-NP was investigated by correlating the P450-dependent catalytic activities in each microsomal sample with the levels of individual metabolites formed by the same microsomes and by examining the effects of agents that can either inhibit or stimulate specific P450 enzymes in mono-NP metabolism. mono-np 359-366 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 10197616-9 1999 Specifically, P450 3A4 was responsible for the formation of 3-hydroxy-1nitropyrene from 1-NP and the formation of trans-9,10-dihydro-9,10dihydroxy-4-nitropyrene, 9(10)-hydroxy-4-nitropyrene, and 4-aminopyrene from 4-NP. 1-nitropyrene-3-ol 60-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 10197616-9 1999 Specifically, P450 3A4 was responsible for the formation of 3-hydroxy-1nitropyrene from 1-NP and the formation of trans-9,10-dihydro-9,10dihydroxy-4-nitropyrene, 9(10)-hydroxy-4-nitropyrene, and 4-aminopyrene from 4-NP. 1-nitropyrene 88-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 10197616-9 1999 Specifically, P450 3A4 was responsible for the formation of 3-hydroxy-1nitropyrene from 1-NP and the formation of trans-9,10-dihydro-9,10dihydroxy-4-nitropyrene, 9(10)-hydroxy-4-nitropyrene, and 4-aminopyrene from 4-NP. trans-9,10-dihydro-9,10dihydroxy-4-nitropyrene 114-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 10197616-9 1999 Specifically, P450 3A4 was responsible for the formation of 3-hydroxy-1nitropyrene from 1-NP and the formation of trans-9,10-dihydro-9,10dihydroxy-4-nitropyrene, 9(10)-hydroxy-4-nitropyrene, and 4-aminopyrene from 4-NP. 9(10)-hydroxy-4-nitropyrene 162-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 10197616-9 1999 Specifically, P450 3A4 was responsible for the formation of 3-hydroxy-1nitropyrene from 1-NP and the formation of trans-9,10-dihydro-9,10dihydroxy-4-nitropyrene, 9(10)-hydroxy-4-nitropyrene, and 4-aminopyrene from 4-NP. 4-Aminopyrene 195-208 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 10197616-9 1999 Specifically, P450 3A4 was responsible for the formation of 3-hydroxy-1nitropyrene from 1-NP and the formation of trans-9,10-dihydro-9,10dihydroxy-4-nitropyrene, 9(10)-hydroxy-4-nitropyrene, and 4-aminopyrene from 4-NP. 4-nonylphenol 214-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 10101147-10 1999 The antibody also was found to immunoinhibit CYP2B6-catalyzed N-demethylation of (S)-mephenytoin in human liver microsomes by 68 to 79%. Mephenytoin 81-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-51 10101147-11 1999 The utility of this antibody for determining human liver microsomal CYP2B6 contribution to the ortho-hydroxylation of methoxychlor was demonstrated. Methoxychlor 118-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 10101149-0 1999 Role of CYP2B6 and CYP3A4 in the in vitro N-dechloroethylation of (R)- and (S)-ifosfamide in human liver microsomes. Nitrogen 42-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 10101149-0 1999 Role of CYP2B6 and CYP3A4 in the in vitro N-dechloroethylation of (R)- and (S)-ifosfamide in human liver microsomes. (r)- and (s)-ifosfamide 66-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-14 10037683-0 1999 The repressed nuclear receptor CAR responds to phenobarbital in activating the human CYP2B6 gene. Phenobarbital 47-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 10037683-1 1999 The endogenous CYP2B6 gene becomes phenobarbital (PB) inducible in androstenol-treated HepG2 cells either transiently or stably transfected with a nuclear receptor CAR expression vector. Phenobarbital 35-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 10037683-1 1999 The endogenous CYP2B6 gene becomes phenobarbital (PB) inducible in androstenol-treated HepG2 cells either transiently or stably transfected with a nuclear receptor CAR expression vector. Phenobarbital 50-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 10037683-1 1999 The endogenous CYP2B6 gene becomes phenobarbital (PB) inducible in androstenol-treated HepG2 cells either transiently or stably transfected with a nuclear receptor CAR expression vector. Androstenols 67-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 10037683-6 1999 Thus, activation of the repressed nuclear receptor CAR appears to be a versatile mediator that regulates PB induction of the CYP2B and other genes. Phenobarbital 105-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-130 10350185-0 1999 Roles of CYP2A6 and CYP2B6 in nicotine C-oxidation by human liver microsomes. Nicotine 30-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-26 10350185-1 1999 Nicotine C-oxidation by recombinant human cytochrome P450 (P450 or CYP) enzymes and by human liver microsomes was investigated using a convenient high-performance liquid chromatographic method. nicotine c 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-70 10350185-6 1999 Contribution of CYP2B6 (as well as CYP2A6) was demonstrated by experiments with the effects of orphenadrine (and also coumarin and anti-CYP2A6) on the nicotine C-oxidation activities by human liver microsomes at 500 microM nicotine. Orphenadrine 95-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 10350185-6 1999 Contribution of CYP2B6 (as well as CYP2A6) was demonstrated by experiments with the effects of orphenadrine (and also coumarin and anti-CYP2A6) on the nicotine C-oxidation activities by human liver microsomes at 500 microM nicotine. coumarin 118-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 10350185-6 1999 Contribution of CYP2B6 (as well as CYP2A6) was demonstrated by experiments with the effects of orphenadrine (and also coumarin and anti-CYP2A6) on the nicotine C-oxidation activities by human liver microsomes at 500 microM nicotine. Nicotine 151-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 10350185-6 1999 Contribution of CYP2B6 (as well as CYP2A6) was demonstrated by experiments with the effects of orphenadrine (and also coumarin and anti-CYP2A6) on the nicotine C-oxidation activities by human liver microsomes at 500 microM nicotine. Nicotine 223-231 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 10051404-1 1999 Sterol 12alpha-hydroxylase (CYP8B1) is a hepatic cytochrome P-450 that controls the ratio of cholic acid over chenodeoxycholic acid in bile and thus controls the solubility of cholesterol. Cholic Acid 93-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-65 10051404-1 1999 Sterol 12alpha-hydroxylase (CYP8B1) is a hepatic cytochrome P-450 that controls the ratio of cholic acid over chenodeoxycholic acid in bile and thus controls the solubility of cholesterol. Cholesterol 176-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-65 10051404-1 1999 Sterol 12alpha-hydroxylase (CYP8B1) is a hepatic cytochrome P-450 that controls the ratio of cholic acid over chenodeoxycholic acid in bile and thus controls the solubility of cholesterol. Chenodeoxycholic Acid 110-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-65 10219963-2 1999 Studies using human liver microsomes and recombinant human cytochrome P450 (P450) enzymes and flavin-containing monooxygenase (FMO) were performed to identify the enzymes responsible for the formation of zotepine metabolites in man. zotepine 204-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-80 10219964-2 1999 Effects of arachidonic acid, prostaglandins, retinol, retinoic acid and cholecalciferol on xenobiotic oxidations catalysed by 12 recombinant human cytochrome P450 (P450 or CYP) enzymes and by human liver microsomes have been investigated. Arachidonic Acid 11-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-175 10219964-2 1999 Effects of arachidonic acid, prostaglandins, retinol, retinoic acid and cholecalciferol on xenobiotic oxidations catalysed by 12 recombinant human cytochrome P450 (P450 or CYP) enzymes and by human liver microsomes have been investigated. Prostaglandins 29-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-175 10219964-2 1999 Effects of arachidonic acid, prostaglandins, retinol, retinoic acid and cholecalciferol on xenobiotic oxidations catalysed by 12 recombinant human cytochrome P450 (P450 or CYP) enzymes and by human liver microsomes have been investigated. Vitamin A 45-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-175 10219964-2 1999 Effects of arachidonic acid, prostaglandins, retinol, retinoic acid and cholecalciferol on xenobiotic oxidations catalysed by 12 recombinant human cytochrome P450 (P450 or CYP) enzymes and by human liver microsomes have been investigated. Tretinoin 54-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-175 10219964-2 1999 Effects of arachidonic acid, prostaglandins, retinol, retinoic acid and cholecalciferol on xenobiotic oxidations catalysed by 12 recombinant human cytochrome P450 (P450 or CYP) enzymes and by human liver microsomes have been investigated. Cholecalciferol 72-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 147-175 9925971-5 1999 For formation of M2, both a rabbit CYP3A polyclonal antibody (110 microliter/mg microsomal protein) and ketoconazole (2 micromol/l), a CYP3A inhibitor, showed about 50% inhibitory effects; other specific inhibitors of CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP2E1 showed no significant effects. Ketoconazole 104-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 226-232 10026272-6 1999 Incubation of ferric P420 nNOS with BH4 alone or BH4 and L-arginine resulted in time-dependent reactivation of catalysis and associated recovery of P450 character. ferric p420 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 10026272-6 1999 Incubation of ferric P420 nNOS with BH4 alone or BH4 and L-arginine resulted in time-dependent reactivation of catalysis and associated recovery of P450 character. sapropterin 36-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 10026272-6 1999 Incubation of ferric P420 nNOS with BH4 alone or BH4 and L-arginine resulted in time-dependent reactivation of catalysis and associated recovery of P450 character. sapropterin 49-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 10026272-6 1999 Incubation of ferric P420 nNOS with BH4 alone or BH4 and L-arginine resulted in time-dependent reactivation of catalysis and associated recovery of P450 character. Arginine 57-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 9888815-0 1999 Dynamics of protein-bound water in the heme domain of P450BM3 studied by high-pressure spectroscopy: comparison with P450cam and P450 2B4. Water 26-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 10022426-10 1999 Steroid-producing cells autoantibodies were found in 11/13 (85%) of patients with idiopathic premature ovarian failure associated with autoimmune Addison"s disease, and autoantibodies to 17alpha-hydroxylase and/or P450 side chain cleavage were found 12/13 (92%) of patients; the only case negative for all these three markers suffered from Turner"s syndrome. Steroids 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 214-218 9929510-3 1999 Methanol did not substantially inhibit (</=10%) any of the activities at 0.3%, but did inhibit CYP1A1, CYP2B6, CYP2C9, and CYP2D6 by 12 to 26% at 1%. Methanol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 9929510-4 1999 In contrast, 0.1% ethanol inhibited CYP1A1, CYP2B6, and CYP2C19 by 20 to 30%. Ethanol 18-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-50 9929510-9 1999 At 1%, acetonitrile decreased activities of CYP1A1 and CYP2B6 by 40 to 60%, and inhibited CYP2A6, CYP3A4, CYP2C19, and CYP2D6 activity by 10 to 20%. acetonitrile 7-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 9918546-10 1999 Furthermore, of 13 human cytochrome P-450s (P-450s) examined, five (CYP1A1, CYP1A2, CYP2B6, CYP2E1, and CYP3A4) were active in the metabolic activation of coumarin, suggesting a potential risk of coumarin toxicity in humans. coumarin 155-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 9918546-10 1999 Furthermore, of 13 human cytochrome P-450s (P-450s) examined, five (CYP1A1, CYP1A2, CYP2B6, CYP2E1, and CYP3A4) were active in the metabolic activation of coumarin, suggesting a potential risk of coumarin toxicity in humans. coumarin 196-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 9888815-0 1999 Dynamics of protein-bound water in the heme domain of P450BM3 studied by high-pressure spectroscopy: comparison with P450cam and P450 2B4. Heme 39-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 10730187-7 1999 Proposed mechanisms generally involve higher oxidation states of the iron (Fe = O), analogous to those for P450, and peroxidase systems. Iron 69-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-127 10027632-0 1999 Highly sensitive high-performance liquid chromatographic assay for coumarin 7-hydroxylation and 7-ethoxycoumarin O-deethylation by human liver cytochrome P450 enzymes. coumarin 67-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 10027632-0 1999 Highly sensitive high-performance liquid chromatographic assay for coumarin 7-hydroxylation and 7-ethoxycoumarin O-deethylation by human liver cytochrome P450 enzymes. 7-ethoxycoumarin 96-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 10027632-1 1999 A highly sensitive method for the determination of coumarin 7-hydroxylation and 7-ethoxycoumarin O-deethylation by human cytochrome P450 (P450 or CYP) enzymes was developed using high-performance liquid chromatography (HPLC). coumarin 51-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-136 10027632-1 1999 A highly sensitive method for the determination of coumarin 7-hydroxylation and 7-ethoxycoumarin O-deethylation by human cytochrome P450 (P450 or CYP) enzymes was developed using high-performance liquid chromatography (HPLC). coumarin 51-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-149 10027632-1 1999 A highly sensitive method for the determination of coumarin 7-hydroxylation and 7-ethoxycoumarin O-deethylation by human cytochrome P450 (P450 or CYP) enzymes was developed using high-performance liquid chromatography (HPLC). 7-ethoxycoumarin 80-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-136 10027632-1 1999 A highly sensitive method for the determination of coumarin 7-hydroxylation and 7-ethoxycoumarin O-deethylation by human cytochrome P450 (P450 or CYP) enzymes was developed using high-performance liquid chromatography (HPLC). 7-ethoxycoumarin 80-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-149 10027632-3 1999 With this high sensitivity, the kinetics of coumarin 7-hydroxylation and 7-ethoxycoumarin O-deethylation catalyzed by human liver microsomal and recombinant P450 enzymes were determined more precisely. coumarin 44-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-161 10027632-3 1999 With this high sensitivity, the kinetics of coumarin 7-hydroxylation and 7-ethoxycoumarin O-deethylation catalyzed by human liver microsomal and recombinant P450 enzymes were determined more precisely. 7-ethoxycoumarin 73-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-161 10447581-3 1999 Recombinant human CYP3A4 and CYP2B6 exhibited catalytic activity with respect to both pathways of trofosfamide. trofosfamide 98-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 9862748-7 1999 These models were then used to predict the Km (apparent) values of a test set of structurally diverse substrates for CYP2B6 not included in the model building, specifically lidocaine, amitriptyline, bupropion, arteether, and verapamil. Lidocaine 173-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 9862748-7 1999 These models were then used to predict the Km (apparent) values of a test set of structurally diverse substrates for CYP2B6 not included in the model building, specifically lidocaine, amitriptyline, bupropion, arteether, and verapamil. Verapamil 225-234 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-123 10697674-3 1999 PAs are metabolized by the liver P450 system to reactive dehydroalkaloid (DHA) intermediates. reactive dehydroalkaloid 48-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 10697674-3 1999 PAs are metabolized by the liver P450 system to reactive dehydroalkaloid (DHA) intermediates. Dihydroalprenolol 74-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 9922992-0 1998 Ethanol interactions with other cytochrome P450 substrates including drugs, xenobiotics, and carcinogens. Ethanol 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 10905564-1 1999 Oxazaphosphorines are inactive anticancer prodrugs that are bioactivated by hepatic cytochrome P450. oxazaphosphorines 0-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-99 10905564-2 1999 Besides hepatic metabolism, there is increasing interest in the possibility of intratumoral activation of oxazaphosphorines by P450. oxazaphosphorines 106-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 10905564-9 1999 In summary, we have demonstrated that tumors of the CNS express P450, indicating that activation of prodrugs like oxazaphosphorines may take place intratumorally. oxazaphosphorines 114-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 9865424-2 1998 Debrisoquine hydroxylase (CYP2D6), one member of the p450 hemoproteins, has polymorphic expression leading to poor metabolism of debrisoquine and similar compounds in approximately 7% of Caucasians. Debrisoquin 129-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 9860924-5 1998 In addition, the location of P450 3A4 residues capable of influencing homotropic stimulation by substrates and heterotropic stimulation by flavonoids has been identified. Flavonoids 139-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 9922992-2 1998 This effect is due primarily to induction by ethanol of a specific cytochrome P450 (CYP2E1) responsible for enhanced oxidation of ethanol and other P450 substrates and, consequently, for metabolic tolerance to these substances. Ethanol 45-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 9922992-2 1998 This effect is due primarily to induction by ethanol of a specific cytochrome P450 (CYP2E1) responsible for enhanced oxidation of ethanol and other P450 substrates and, consequently, for metabolic tolerance to these substances. Ethanol 45-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 9922992-2 1998 This effect is due primarily to induction by ethanol of a specific cytochrome P450 (CYP2E1) responsible for enhanced oxidation of ethanol and other P450 substrates and, consequently, for metabolic tolerance to these substances. Ethanol 130-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 9922992-2 1998 This effect is due primarily to induction by ethanol of a specific cytochrome P450 (CYP2E1) responsible for enhanced oxidation of ethanol and other P450 substrates and, consequently, for metabolic tolerance to these substances. Ethanol 130-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 9922992-6 1998 The metabolism of steroids and vitamins is catalyzed by P450 and is altered in chronic alcoholics. Steroids 18-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 9756035-0 1998 Identification of the human P-450 enzymes responsible for the sulfoxidation and thiono-oxidation of diethyldithiocarbamate methyl ester: role of P-450 enzymes in disulfiram bioactivation. methyl diethyldithiocarbamate 100-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-33 9863553-5 1998 P450 3A is the most abundant P450 protein in the adult human liver and is able to transform hundreds of substrates into either drugs or endogenous compounds such as testosterone. Testosterone 165-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 9863553-5 1998 P450 3A is the most abundant P450 protein in the adult human liver and is able to transform hundreds of substrates into either drugs or endogenous compounds such as testosterone. Testosterone 165-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 9863553-8 1998 Erythromycin metabolism and its P450 effects are used to illustrate the complexity and the consequences of metabolic transformation of a given drug. Erythromycin 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 9766669-4 1998 Intratumoral cytochrome P450 expression conferred substantial sensitivity to CPA cytotoxicity, with the most dramatic effects seen with CYP2B6. Cyclophosphamide 77-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-142 9766669-10 1998 CYP2B6 plus P450 reductase and CYP2C18-Met plus P450 reductase thus appear to be excellent gene combinations for use with CPA in P450/prodrug activation-based cancer gene therapy. Cyclophosphamide 122-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 9825830-0 1998 In-vitro characterization of the cytochrome P450 isoenzymes involved in the back oxidation and N-dealkylation of reduced haloperidol. Haloperidol 121-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 9825830-1 1998 In-vitro studies were performed using human liver microsomes and c-DNA-expressed human P450 isoforms to identify the cytochrome P450 isoenzyme(s) involved in the back oxidation and N-dealkylation of reduced haloperidol. Haloperidol 207-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 9756035-0 1998 Identification of the human P-450 enzymes responsible for the sulfoxidation and thiono-oxidation of diethyldithiocarbamate methyl ester: role of P-450 enzymes in disulfiram bioactivation. methyl diethyldithiocarbamate 100-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-150 9756035-3 1998 Several approaches were used in an attempt to delineate the particular P-450 enzyme(s) involved in the sulfoxidation and thiono-oxidation of DDTC-Me. methyl diethyldithiocarbamate 141-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-76 9756035-5 1998 Multiple human P-450 enzymes (CYP3A4, CYP1A2, CYP2A6, and CYP2D6) catalyzed the sulfoxidation of DDTC-Me, as determined with cDNA-expressed enzymes. methyl diethyldithiocarbamate 97-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-20 9806171-4 1998 Among the cytochrome P450 enzymes capable of metabolizing nitrosamines, CYP2A6, CYP2E1 and CYP2B6 are expressed in BECs. Nitrosamines 58-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-97 9734472-2 1998 It has been proposed that metabolites of arachidonic acid (AA), formed by cytochrome P-450 monooxygenase (P450), are endothelium-derived hyperpolarizing factors (EDHFs). Arachidonic Acid 41-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-110 9734472-2 1998 It has been proposed that metabolites of arachidonic acid (AA), formed by cytochrome P-450 monooxygenase (P450), are endothelium-derived hyperpolarizing factors (EDHFs). edhfs 162-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-110 9734472-12 1998 Both 17-ODYA and miconazole (10(-5) mol/L, a chemically distinct P450 inhibitor) further reduced the dilation to AA in the presence of INDO. 17-octadecynoic acid 5-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 9734472-12 1998 Both 17-ODYA and miconazole (10(-5) mol/L, a chemically distinct P450 inhibitor) further reduced the dilation to AA in the presence of INDO. Miconazole 17-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 9734472-12 1998 Both 17-ODYA and miconazole (10(-5) mol/L, a chemically distinct P450 inhibitor) further reduced the dilation to AA in the presence of INDO. Indomethacin 135-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 9758440-11 1998 Finally, the mean 17-OHP level in patients with LH levels which induced marked P450 down-regulation (i.e. more than 12 IU/l) was similar to that in patients with LH levels within the normal range (i.e. less than 6 IU/l). 17-alpha-Hydroxyprogesterone 18-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 9733665-4 1998 Incubation of methoxychlor with microsomes from insect cells overexpressing either CYP1A2, CYP2C18, or CYP2C19 yielded mono-OH-M with turnover numbers of 14.9, 15.5, and 39.1 nmol/min/nmol of P450, respectively. Methoxychlor 14-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-196 9758440-11 1998 Finally, the mean 17-OHP level in patients with LH levels which induced marked P450 down-regulation (i.e. more than 12 IU/l) was similar to that in patients with LH levels within the normal range (i.e. less than 6 IU/l). Luteinizing Hormone 48-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 9733666-1 1998 The substrate structure-activity relationships described for the major human drug-metabolizing cytochrome P450 (P450 or CYP) enzymes suggest that the H1 receptor antagonist terfenadine could interact with CYP2D6 either as a substrate or as an inhibitor, in addition to its known ability to act as a substrate for CYP3A4. Terfenadine 173-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-110 9733666-1 1998 The substrate structure-activity relationships described for the major human drug-metabolizing cytochrome P450 (P450 or CYP) enzymes suggest that the H1 receptor antagonist terfenadine could interact with CYP2D6 either as a substrate or as an inhibitor, in addition to its known ability to act as a substrate for CYP3A4. Terfenadine 173-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-123 9758440-14 1998 However, in contrast to in vitro studies, LH levels which induce P450 down-regulation appear to be less effective on delta(4)17,20-lyase than on delta(4)17-hydroxylase in HW. Luteinizing Hormone 42-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 9744576-1 1998 Mono-specific antibodies against the human cytochrome P450 (P450) enzymes CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2B6, CYP2D6, CYP2E1, CYP3A4, CYP3A5 and CYP4A11 and an antibody that binds to CYP2C8, CYP2C9 and CYP2C19 have been produced by immunising rabbits with synthetic peptides representing small regions of each of these P450 enzymes. Peptides 270-278 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 9733666-8 1998 The formation of the hydroxylated metabolite (hydroxyterfenadine) in microsomes prepared from human liver and specific P450 cDNA-transfected B lymphoblastoid cells indicated that only CYP2D6 and CYP3A4 were involved in this transformation. MDL-17523 46-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-123 9744576-1 1998 Mono-specific antibodies against the human cytochrome P450 (P450) enzymes CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2B6, CYP2D6, CYP2E1, CYP3A4, CYP3A5 and CYP4A11 and an antibody that binds to CYP2C8, CYP2C9 and CYP2C19 have been produced by immunising rabbits with synthetic peptides representing small regions of each of these P450 enzymes. Peptides 270-278 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-64 9732386-5 1998 CYP2B6 levels were found to significantly (P < .0001) correlate with S-mephenytoin N-demethylation to nirvanol (r2 = 0.89), 7-hydroxy-4-trifluoromethylcoumarin formation (r2 = 0.81) and several markers of CYP3A levels and activity. Mephenytoin 72-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 9732386-5 1998 CYP2B6 levels were found to significantly (P < .0001) correlate with S-mephenytoin N-demethylation to nirvanol (r2 = 0.89), 7-hydroxy-4-trifluoromethylcoumarin formation (r2 = 0.81) and several markers of CYP3A levels and activity. ethylphenylhydantoin 105-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 9732386-5 1998 CYP2B6 levels were found to significantly (P < .0001) correlate with S-mephenytoin N-demethylation to nirvanol (r2 = 0.89), 7-hydroxy-4-trifluoromethylcoumarin formation (r2 = 0.81) and several markers of CYP3A levels and activity. 7-hydroxy-4-trifluoromethylcoumarin 127-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 9732386-8 1998 Testosterone 16beta-hydroxylation by expressed CYP2B6 resulted in atypical kinetics characteristic of substrate activation. Testosterone 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 9732386-10 1998 In conclusion, the highly specific MAb 49-10-20 was used to provide further confirmation that S-mephenytoin N-demethylation to nirvanol is a CYP2B6 selective probe. Mephenytoin 94-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 9732386-10 1998 In conclusion, the highly specific MAb 49-10-20 was used to provide further confirmation that S-mephenytoin N-demethylation to nirvanol is a CYP2B6 selective probe. ethylphenylhydantoin 127-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-147 9698292-5 1998 Of the recombinant P450 isoforms tested, only CYP2B6 and CYP2C9 formed nirvanol from (S)-MP. ethylphenylhydantoin 71-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 9698292-9 1998 Therefore, the N-demethylation of (S)-MP to nirvanol may be a useful means of probing the activity of CYP2B6 in vitro when concentrations of >1000 microM are used, but it is unlikely to be a suitable phenotyping tool for this isoform in vivo, where concentrations of >1000 microM are rarely encountered. Nitrogen 15-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 9698292-9 1998 Therefore, the N-demethylation of (S)-MP to nirvanol may be a useful means of probing the activity of CYP2B6 in vitro when concentrations of >1000 microM are used, but it is unlikely to be a suitable phenotyping tool for this isoform in vivo, where concentrations of >1000 microM are rarely encountered. 6-trimethylsilylthio-9-trimethylsilylpurine 34-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 9698292-9 1998 Therefore, the N-demethylation of (S)-MP to nirvanol may be a useful means of probing the activity of CYP2B6 in vitro when concentrations of >1000 microM are used, but it is unlikely to be a suitable phenotyping tool for this isoform in vivo, where concentrations of >1000 microM are rarely encountered. ethylphenylhydantoin 44-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 9711991-0 1998 The induction of P450-mediated oxidation of all-trans retinoic acid by retinoids in head and neck squamous cell carcinoma cell lines. all-trans 44-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 9711991-4 1998 It was found that the enhanced catabolism brought on by P450 induction was blocked when RA was added to AMC-HN-6 along with actinomycin D or cyclohexamide. Tretinoin 88-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 9690701-0 1998 Appetite suppressant drugs as inhibitors of human cytochromes P450: in vitro inhibition of P450-2D6 by D- and L-fenfluramine, but not phentermine. d- and l-fenfluramine 103-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 9690701-0 1998 Appetite suppressant drugs as inhibitors of human cytochromes P450: in vitro inhibition of P450-2D6 by D- and L-fenfluramine, but not phentermine. d- and l-fenfluramine 103-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 9690701-1 1998 The activity of D-fenfluramine, L-fenfluramine, and phentermine as inhibitors of five human cytochromes P450 was evaluated using human liver microsomes in vitro. Dexfenfluramine 16-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-108 9690701-1 1998 The activity of D-fenfluramine, L-fenfluramine, and phentermine as inhibitors of five human cytochromes P450 was evaluated using human liver microsomes in vitro. Fenfluramine 32-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-108 9690701-1 1998 The activity of D-fenfluramine, L-fenfluramine, and phentermine as inhibitors of five human cytochromes P450 was evaluated using human liver microsomes in vitro. Phentermine 52-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-108 9690701-4 1998 However, D- and L-fenfluramine significantly inhibited P450-2D6 activity as measured by dextromethorphan O-demethylation, with mean 50% inhibitory concentrations (15.1 microM) within one order of magnitude of that for fluoxetine (2.7 microM). d- and l-fenfluramine 9-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 9690701-4 1998 However, D- and L-fenfluramine significantly inhibited P450-2D6 activity as measured by dextromethorphan O-demethylation, with mean 50% inhibitory concentrations (15.1 microM) within one order of magnitude of that for fluoxetine (2.7 microM). Dextromethorphan 88-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 9711991-4 1998 It was found that the enhanced catabolism brought on by P450 induction was blocked when RA was added to AMC-HN-6 along with actinomycin D or cyclohexamide. Dactinomycin 124-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 9711991-0 1998 The induction of P450-mediated oxidation of all-trans retinoic acid by retinoids in head and neck squamous cell carcinoma cell lines. Tretinoin 54-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 9711991-4 1998 It was found that the enhanced catabolism brought on by P450 induction was blocked when RA was added to AMC-HN-6 along with actinomycin D or cyclohexamide. 4-[2-(3,5-dimethyl-2-oxocyclohexyl)-2-hydroxyethyl]piperidine-2,6-dione 141-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 9711991-0 1998 The induction of P450-mediated oxidation of all-trans retinoic acid by retinoids in head and neck squamous cell carcinoma cell lines. Retinoids 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-21 9711991-6 1998 P450-mediated oxidation was detectable within 4 hours of RA treatment, and RA catabolism reached its maximum 16 hours after treatment. Tretinoin 57-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 9711991-1 1998 All-trans retinoic acid (RA) can be catabolized into polar metabolites by cytochrome P450 (P450) in several tissues including the skin. Tretinoin 0-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 9711991-7 1998 P450 was induced by 13-cis-RA, 9-cis-RA, and retinal; however, retinol could not induce P450. Isotretinoin 20-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 9711991-7 1998 P450 was induced by 13-cis-RA, 9-cis-RA, and retinal; however, retinol could not induce P450. Alitretinoin 31-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 9711991-1 1998 All-trans retinoic acid (RA) can be catabolized into polar metabolites by cytochrome P450 (P450) in several tissues including the skin. Tretinoin 0-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 9711991-8 1998 In conclusion, P450 can be induced by retinoids in head and neck SCC (HNSCC) cells and the ability of retinoids to induce P450 can serve as an important factor in determining the biological effect of retinoids. Retinoids 38-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 9711991-8 1998 In conclusion, P450 can be induced by retinoids in head and neck SCC (HNSCC) cells and the ability of retinoids to induce P450 can serve as an important factor in determining the biological effect of retinoids. Retinoids 102-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-126 9711991-8 1998 In conclusion, P450 can be induced by retinoids in head and neck SCC (HNSCC) cells and the ability of retinoids to induce P450 can serve as an important factor in determining the biological effect of retinoids. Retinoids 102-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-126 9711991-1 1998 All-trans retinoic acid (RA) can be catabolized into polar metabolites by cytochrome P450 (P450) in several tissues including the skin. Tretinoin 25-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 9711991-1 1998 All-trans retinoic acid (RA) can be catabolized into polar metabolites by cytochrome P450 (P450) in several tissues including the skin. Tretinoin 25-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 9711991-2 1998 We examined eight different squamous cell carcinoma (SCC) cell lines to determine their capacity to induce P450-mediated oxidation of RA. Tretinoin 134-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-111 9633999-1 1998 The human cytochrome P450 2B6 metabolizes, among numerous other substrates, diazepam, 7-ethoxycoumarin, testosterone, and phenanthrene. Diazepam 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-29 9755460-0 1998 [Inhibition of cytochrome P450 by nitric oxide]. Nitric Oxide 34-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 9755460-2 1998 Although NO reversibly interacts with the heme-iron of P450, the pathophysiological role of this interaction remains to be elucidated. Heme 42-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 9755460-2 1998 Although NO reversibly interacts with the heme-iron of P450, the pathophysiological role of this interaction remains to be elucidated. Iron 47-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-59 9755460-11 1998 These results suggested that NO might form dissociable complexes with the heme moiety of P450 and irreversibly inactivate them. Heme 74-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-93 15012237-1 1998 Cytochrome P450-dependent monooxygenases are a large group of heme-containing enzymes, most of which catalyze NADPH- and O2-dependent hydroxylation reactions. Heme 62-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 15012237-1 1998 Cytochrome P450-dependent monooxygenases are a large group of heme-containing enzymes, most of which catalyze NADPH- and O2-dependent hydroxylation reactions. NADP 110-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 15012237-1 1998 Cytochrome P450-dependent monooxygenases are a large group of heme-containing enzymes, most of which catalyze NADPH- and O2-dependent hydroxylation reactions. Oxygen 121-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 9616194-1 1998 The metabolism of the antidepressant drug trazodone to its active metabolite, m-chlorophenylpiperazine (mCPP), was studied in vitro using human liver microsomal preparations and cDNA-expressed human cytochrome P450 (P450) enzymes. Trazodone 42-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-221 9616194-1 1998 The metabolism of the antidepressant drug trazodone to its active metabolite, m-chlorophenylpiperazine (mCPP), was studied in vitro using human liver microsomal preparations and cDNA-expressed human cytochrome P450 (P450) enzymes. 1-(3-chlorophenyl)piperazine 78-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-221 9618440-0 1998 Metabolism of arachidonic acid to 20-hydroxy-5,8,11, 14-eicosatetraenoic acid by P450 enzymes in human liver: involvement of CYP4F2 and CYP4A11. Arachidonic Acid 14-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 9618440-0 1998 Metabolism of arachidonic acid to 20-hydroxy-5,8,11, 14-eicosatetraenoic acid by P450 enzymes in human liver: involvement of CYP4F2 and CYP4A11. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 34-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 9618440-1 1998 20-Hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE) is a principal arachidonic acid (AA) metabolite formed via P450-dependent oxidation in hepatic and renal microsomes. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 0-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 9618440-1 1998 20-Hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE) is a principal arachidonic acid (AA) metabolite formed via P450-dependent oxidation in hepatic and renal microsomes. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 44-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 9618440-1 1998 20-Hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE) is a principal arachidonic acid (AA) metabolite formed via P450-dependent oxidation in hepatic and renal microsomes. Arachidonic Acid 68-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 9695717-2 1998 A single dose of disulfiram, which significantly diminishes human P450 2E1 activity in vivo, has been used to investigate the role of CYP2E1 in human drug metabolism and to prevent CYP2E1-mediated biotransformation. Disulfiram 17-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 9695717-12 1998 When single-dose disulfiram is used as an in vivo probe for P450, inhibition of drug metabolism suggests involvement of CYP2E1 but not CYP2A6. Disulfiram 17-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-64 9625734-1 1998 Of seven cDNA-expressed human cytochrome P450 (P450) enzymes (P450s 1A2, 2B6, 2C9, 2C19, 2D6, 2E1, and 3A4) examined, P450 1A2 was the most active in catalyzing 2- and 4-hydroxylations of estradiol and estrone. Estradiol 188-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-51 9625734-1 1998 Of seven cDNA-expressed human cytochrome P450 (P450) enzymes (P450s 1A2, 2B6, 2C9, 2C19, 2D6, 2E1, and 3A4) examined, P450 1A2 was the most active in catalyzing 2- and 4-hydroxylations of estradiol and estrone. Estrone 202-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-51 9633999-1 1998 The human cytochrome P450 2B6 metabolizes, among numerous other substrates, diazepam, 7-ethoxycoumarin, testosterone, and phenanthrene. 7-ethoxycoumarin 86-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-29 9633999-1 1998 The human cytochrome P450 2B6 metabolizes, among numerous other substrates, diazepam, 7-ethoxycoumarin, testosterone, and phenanthrene. Testosterone 104-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-29 9633999-1 1998 The human cytochrome P450 2B6 metabolizes, among numerous other substrates, diazepam, 7-ethoxycoumarin, testosterone, and phenanthrene. phenanthrene 122-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-29 9630846-0 1998 Covalent binding of carbamazepine reactive metabolites to P450 isoforms present in the skin. Carbamazepine 20-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 9630846-2 1998 Upon metabolism by cytochrome P450, carbamazepine may produce reactive metabolites. Carbamazepine 36-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 9630846-4 1998 Carbamazepine reactive metabolites covalent binding to human liver microsomes involved P450 subfamilies 1A, 2C and 3A. Carbamazepine 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 9630846-5 1998 Specific covalent binding to yeasts expressing different P450s showed that carbamazepine reactive metabolites bound specifically to P450 1A2 and 3A4. Carbamazepine 75-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 9630846-7 1998 Consequently, the production in epidermis of carbamazepine reactive metabolites is theoretically possible with formation of P450 adduct metabolites. Carbamazepine 45-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 9589661-1 1998 The physiological importance of adrenal 21-hydroxylase cytochrome P450 (CYP21) expression is clearly demonstrated by 21-hydroxylase deficiency, which results in adrenal hyperplasia and over-production of C19 steroids, leading to virilization. c19 steroids 204-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 9518732-5 1998 Bath administration of the imidazole antimycotics, clotrimazole, econazole and miconazole, which are potent P450 inhibitors, significantly suppressed cardiac ICa. imidazole 27-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 9518732-5 1998 Bath administration of the imidazole antimycotics, clotrimazole, econazole and miconazole, which are potent P450 inhibitors, significantly suppressed cardiac ICa. Clotrimazole 51-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 9518732-5 1998 Bath administration of the imidazole antimycotics, clotrimazole, econazole and miconazole, which are potent P450 inhibitors, significantly suppressed cardiac ICa. Econazole 65-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 9518732-5 1998 Bath administration of the imidazole antimycotics, clotrimazole, econazole and miconazole, which are potent P450 inhibitors, significantly suppressed cardiac ICa. Miconazole 79-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-112 9518732-11 1998 In addition, intracellular dialysis with 2 mM cAMP abolished the P450 inhibitor-induced suppression of ICa. Cyclic AMP 46-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 9518732-11 1998 In addition, intracellular dialysis with 2 mM cAMP abolished the P450 inhibitor-induced suppression of ICa. isocyanic acid 103-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-69 9518732-20 1998 Extracellular administration of 11, 12-epoxyeicosatrienoic acid, one of the P450-mediated metabolites of arachidonic acid, enhanced ICa and intracellular cAMP content. 11,12-epoxy-5,8,14-eicosatrienoic acid 32-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 9518732-20 1998 Extracellular administration of 11, 12-epoxyeicosatrienoic acid, one of the P450-mediated metabolites of arachidonic acid, enhanced ICa and intracellular cAMP content. Arachidonic Acid 105-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 9518732-20 1998 Extracellular administration of 11, 12-epoxyeicosatrienoic acid, one of the P450-mediated metabolites of arachidonic acid, enhanced ICa and intracellular cAMP content. isocyanic acid 132-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 9518732-20 1998 Extracellular administration of 11, 12-epoxyeicosatrienoic acid, one of the P450-mediated metabolites of arachidonic acid, enhanced ICa and intracellular cAMP content. Cyclic AMP 154-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 9518732-21 1998 The epoxyeicosatrienoic acid also restored the amplitude of the reduced ICa in P450 antibody-dialysed myocytes. epoxyeicosatrienoic acid 4-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 9518732-21 1998 The epoxyeicosatrienoic acid also restored the amplitude of the reduced ICa in P450 antibody-dialysed myocytes. isocyanic acid 72-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 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 2 subfamily B member 6 Homo sapiens 41-45 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 2 subfamily B member 6 Homo sapiens 41-45 9589661-11 1998 Inhibition of protein synthesis with cycloheximide blocked induction of CYP21 as well as type II 3 beta-hydroxysteroid dehydrogenase (3 beta HSDII) mRNA expression in response to AII, forskolin, and dibutyryl cAMP, but had no effect on 17 alpha-hydroxylase cytochrome P450 (CYP17) or cholesterol side-chain cleavage cytochrome P450 (CYP11A) mRNA. Cycloheximide 37-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 268-272 9589661-11 1998 Inhibition of protein synthesis with cycloheximide blocked induction of CYP21 as well as type II 3 beta-hydroxysteroid dehydrogenase (3 beta HSDII) mRNA expression in response to AII, forskolin, and dibutyryl cAMP, but had no effect on 17 alpha-hydroxylase cytochrome P450 (CYP17) or cholesterol side-chain cleavage cytochrome P450 (CYP11A) mRNA. Cycloheximide 37-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 327-331 9653975-2 1998 ETFMC was incubated at 37 degrees C with human hepatic microsomes or microsomes prepared from a lymphoblastoid cell line that expresses human CYP2B6. 7-ethoxy-4-trifluoromethylcoumarin 0-5 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-148 9531517-11 1998 These results demonstrate that CYP3A4 is the primary isozyme involved in the metabolism of AE to its active metabolite, DQHS, with secondary contributions by CYP2B6 and -3A5. artemotil 91-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-164 9394031-1 1997 The P450 2A6 catalyzed 7-hydroxylation of coumarin proceeded with a mean Km of 0.40 (+/-0.13) microM and Vmax of 6.34 nmol/nmol P450/min (36-fold variation) in microsomal preparations from a panel of 12 human livers. coumarin 42-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-136 9531515-7 1998 Several recombinant human P450 enzymes were incubated with [1,2,6,7-3H]testosterone, and only cDNA-expressed CYP3A4 catalyzed a high rate of tritium release. [1,2,6,7-3h]testosterone 59-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 9531515-7 1998 Several recombinant human P450 enzymes were incubated with [1,2,6,7-3H]testosterone, and only cDNA-expressed CYP3A4 catalyzed a high rate of tritium release. Tritium 141-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 9531515-9 1998 However, the tritium-release assay may have limited value in measuring CYP3A activity in liver microsomes from other species due to the presence of other P450 enzymes that can catalyze tritium release from [1,2,6,7-3H]testosterone. Tritium 13-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 9531515-9 1998 However, the tritium-release assay may have limited value in measuring CYP3A activity in liver microsomes from other species due to the presence of other P450 enzymes that can catalyze tritium release from [1,2,6,7-3H]testosterone. Tritium 185-192 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 9531515-9 1998 However, the tritium-release assay may have limited value in measuring CYP3A activity in liver microsomes from other species due to the presence of other P450 enzymes that can catalyze tritium release from [1,2,6,7-3H]testosterone. [1,2,6,7-3h]testosterone 206-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 9531517-10 1998 CYP3A4 metabolized AE to dihydroqinghaosu at a rate approximately 10 times that of CYP2B6 and approximately 4.5-fold greater than that of CYP3A5. artemotil 19-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 9586947-0 1998 Role of human liver P450s and cytochrome b5 in the reductive metabolism of 3"-azido-3"-deoxythymidine (AZT) to 3"-amino-3"-deoxythymidine. Zidovudine 75-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 9586947-0 1998 Role of human liver P450s and cytochrome b5 in the reductive metabolism of 3"-azido-3"-deoxythymidine (AZT) to 3"-amino-3"-deoxythymidine. Zidovudine 103-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 9586947-0 1998 Role of human liver P450s and cytochrome b5 in the reductive metabolism of 3"-azido-3"-deoxythymidine (AZT) to 3"-amino-3"-deoxythymidine. 3'-aminothymidine 111-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 9586947-1 1998 Our laboratory has shown that human liver microsomes metabolize the anti-HIV drug 3"-azido-3"-deoxythymidine (AZT) via a P450-type reductive reaction to a toxic metabolite 3"-amino-3"-deoxythymidine (AMT). Zidovudine 82-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 9586947-1 1998 Our laboratory has shown that human liver microsomes metabolize the anti-HIV drug 3"-azido-3"-deoxythymidine (AZT) via a P450-type reductive reaction to a toxic metabolite 3"-amino-3"-deoxythymidine (AMT). Zidovudine 110-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 9586947-1 1998 Our laboratory has shown that human liver microsomes metabolize the anti-HIV drug 3"-azido-3"-deoxythymidine (AZT) via a P450-type reductive reaction to a toxic metabolite 3"-amino-3"-deoxythymidine (AMT). 3'-aminothymidine 172-198 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 9586947-1 1998 Our laboratory has shown that human liver microsomes metabolize the anti-HIV drug 3"-azido-3"-deoxythymidine (AZT) via a P450-type reductive reaction to a toxic metabolite 3"-amino-3"-deoxythymidine (AMT). 3'-aminothymidine 200-203 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 9586947-4 1998 Involvement of a specific P450 enzyme in AZT reduction was not detected by using human P450 substrates and inhibitors. Zidovudine 41-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 9544620-5 1998 Two other metabolites were the cytochrome P450-mediated (P450) oxidation products 2-amino-8-(hydroxymethyl)-3-methylimidazo[4,5-f]quinoxaline (8-CH2OH-MeIQx), and N2-(beta-1-glucosiduronyl)-N-hydroxy-2-amino-3,8-dimethylimidaz o[4,5-f]quinoxaline (NOH-MeIQx-N2-Gl). 3H-Imidazo(4,5-f)quinoxaline-8-methanol, 2-amino-3-methyl- 82-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-61 9544620-5 1998 Two other metabolites were the cytochrome P450-mediated (P450) oxidation products 2-amino-8-(hydroxymethyl)-3-methylimidazo[4,5-f]quinoxaline (8-CH2OH-MeIQx), and N2-(beta-1-glucosiduronyl)-N-hydroxy-2-amino-3,8-dimethylimidaz o[4,5-f]quinoxaline (NOH-MeIQx-N2-Gl). 8-ch2oh-meiqx 143-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-61 9544620-5 1998 Two other metabolites were the cytochrome P450-mediated (P450) oxidation products 2-amino-8-(hydroxymethyl)-3-methylimidazo[4,5-f]quinoxaline (8-CH2OH-MeIQx), and N2-(beta-1-glucosiduronyl)-N-hydroxy-2-amino-3,8-dimethylimidaz o[4,5-f]quinoxaline (NOH-MeIQx-N2-Gl). n2-(beta-1-glucosiduronyl)-n-hydroxy-2-amino-3,8-dimethylimidaz o[4,5-f]quinoxaline 163-246 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-61 9544620-5 1998 Two other metabolites were the cytochrome P450-mediated (P450) oxidation products 2-amino-8-(hydroxymethyl)-3-methylimidazo[4,5-f]quinoxaline (8-CH2OH-MeIQx), and N2-(beta-1-glucosiduronyl)-N-hydroxy-2-amino-3,8-dimethylimidaz o[4,5-f]quinoxaline (NOH-MeIQx-N2-Gl). noh-meiqx-n2-gl 248-263 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-61 18406240-2 1998 In this article, we present evidence from molecular genetic studies for involvement of the steroid synthesis gene CYP11a (coding for P450 cholesterol side-chain cleavage) in the aetiology of hyperandrogenism. Steroids 91-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 18406240-2 1998 In this article, we present evidence from molecular genetic studies for involvement of the steroid synthesis gene CYP11a (coding for P450 cholesterol side-chain cleavage) in the aetiology of hyperandrogenism. Cholesterol 138-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 9586947-7 1998 Indeed, AZT reduction among six human liver samples correlated strongly with microsomal b5 content (r2 = 0.96) as well as with aggregate P450 content (r2 = 0.97). Zidovudine 8-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-141 9586947-10 1998 Our results indicate that AZT reduction to AMT by human liver microsomes involves both b5 and P450 enzymes plus their corresponding reductases. Zidovudine 26-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-98 10335404-0 1998 [Chemical synthesis of cytochrome P-450-dependent metabolites of arachidonic acid]. Arachidonic Acid 65-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-39 10335404-1 1998 Approaches to the chemical synthesis of cytochrome P-450-dependent metabolites of arachidonic acid and the biological role of novel metabolites in the arachidonic acid cascade are discussed. Arachidonic Acid 82-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-56 10335404-1 1998 Approaches to the chemical synthesis of cytochrome P-450-dependent metabolites of arachidonic acid and the biological role of novel metabolites in the arachidonic acid cascade are discussed. Arachidonic Acid 151-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-56 14577219-0 1998 Determination of the CYP2B6 component of 7-ethoxy-4-trifluoromethylcoumarin O-deethylation activity in human liver microsomes. 7-ethoxy-4-trifluoromethylcoumarin 41-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 9434738-1 1997 In previous studies, we found that the ascorbic acid (AsA) deficiency caused changes in the amounts of the various forms of cytochrome P450 (P450) in liver microsomes from guinea pigs in a form-specific manner. Ascorbic Acid 39-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-139 9434738-1 1997 In previous studies, we found that the ascorbic acid (AsA) deficiency caused changes in the amounts of the various forms of cytochrome P450 (P450) in liver microsomes from guinea pigs in a form-specific manner. Ascorbic Acid 39-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 9434738-1 1997 In previous studies, we found that the ascorbic acid (AsA) deficiency caused changes in the amounts of the various forms of cytochrome P450 (P450) in liver microsomes from guinea pigs in a form-specific manner. Ascorbic Acid 54-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-139 9434738-1 1997 In previous studies, we found that the ascorbic acid (AsA) deficiency caused changes in the amounts of the various forms of cytochrome P450 (P450) in liver microsomes from guinea pigs in a form-specific manner. Ascorbic Acid 54-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 9434738-11 1997 These results support the idea that the transcription of P450 is regulated by AsA in guinea pigs. Ascorbic Acid 78-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 9416970-3 1997 The correlation coefficient between R-warfarin 7-hydroxylation and 7-ethoxyresorufin O-deethylation activities was 0.73 in these human samples, suggesting that R- and S-warfarin enantiomers are catalyzed by different forms of human cytochrome P450 (P450 or CYP) enzymes. Warfarin 36-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 249-260 11360624-1 1997 With specific designed primers, CYP2B6 and CYP1A1 cDNA were generated by reverse transcription-polymerase chain reaction(RT-PCR) technique performed on total RNAs isolated from human liver and 3-methylcholanthrene(3-MC) induced human amnion FL cells. Methylcholanthrene 193-213 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 9455828-7 1997 We review evidence for an important role for the insulin gene minisatellite in the aetiology of anovulatory PCOS and for the gene coding for P450 cholesterol side chain cleavage (CYP11a) in the mechanism of excessive androgen secretion in women with polycystic ovaries. Cholesterol 146-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 9328296-1 1997 Roles of human cytochrome P450 (P450 or CYP) 2C9, 2C19, and 3A4 in the oxidation of progesterone and testosterone were studied in recombinant P450 enzymes and in human liver microsomes. Progesterone 84-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-48 9328296-1 1997 Roles of human cytochrome P450 (P450 or CYP) 2C9, 2C19, and 3A4 in the oxidation of progesterone and testosterone were studied in recombinant P450 enzymes and in human liver microsomes. Testosterone 101-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-48 11360624-1 1997 With specific designed primers, CYP2B6 and CYP1A1 cDNA were generated by reverse transcription-polymerase chain reaction(RT-PCR) technique performed on total RNAs isolated from human liver and 3-methylcholanthrene(3-MC) induced human amnion FL cells. Methylcholanthrene 214-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 9360635-8 1997 Subsequent introduction of the lacI gene into IIB1 cells resulted in correct transcriptional repression of the lacZ gene that could be alleviated by IPTG, an allosteric inducer of lacI repression. Isopropyl Thiogalactoside 149-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-50 9315688-1 1997 A mutation at the surface of the substrate access channel which dramatically decreases the affinity for some fatty acids in P450(BM-3) was discovered by random mutagenesis. Fatty Acids 109-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-133 9206005-4 1997 In addition to these enzymes, P450 monooxygenases in some fungi were implicated in the degradation of PAHs. Polycyclic Aromatic Hydrocarbons 102-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 9201961-7 1997 Cytochrome P-420cam is produced from substrate-free cytochrome P-450cam in two ways: (i) by temperature elevation up to 60 degrees C and (ii) by exposure to KSCN up to 1.5 M. The secondary structure composition is determined for each temperature and KSCN concentration and compared with the changes observed in the iron ligand CO stretch vibration bands appearing between 1900 and 2000 cm-1. potassium thiocyanate 157-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-68 9316851-6 1997 Enzyme activity varied 23-fold among 15 human liver microsome samples and correlated with CYP2A6-catalyzed coumarin hydroxylase (r2 = 0.85, P < .01) and CYP2B6-catalyzed 7-ethoxytrifluoromethylcoumarin O-deethylase (r2 = 0.82, P < .01) activities. 7-ethoxytrifluoromethylcoumarin 173-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 156-162 9280407-7 1997 CYP2B6-expressing cells were found to be more sensitive than control cells to the cytotoxicity and mutagenicity of cyclophosphamide, aflatoxin B1, and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Cyclophosphamide 115-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 9280407-7 1997 CYP2B6-expressing cells were found to be more sensitive than control cells to the cytotoxicity and mutagenicity of cyclophosphamide, aflatoxin B1, and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Aflatoxin B1 133-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 9280407-7 1997 CYP2B6-expressing cells were found to be more sensitive than control cells to the cytotoxicity and mutagenicity of cyclophosphamide, aflatoxin B1, and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. 4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone 151-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 9224778-0 1997 In vitro identification of the P450 enzymes responsible for the metabolism of ropinirole. ropinirole 78-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 9224778-1 1997 The in vitro metabolism of ropinirole was investigated with the aim of identifying the cytochrome P450 enzymes responsible for its biotransformation. ropinirole 27-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 9224778-8 1997 Multivariate correlation data were consistent with the involvement of the cytochrome P450 enzymes 1A2 and 3A in the metabolism of ropinirole. ropinirole 130-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 9224778-9 1997 Thus, it could be concluded that the major P450 enzyme responsible for ropinirole metabolism at lower (clinically relevant) concentrations is CYP1A2 with a contribution from CYP3A, particularly at higher concentrations. ropinirole 71-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 9253143-12 1997 At the higher concentration (1.8 mM) of styrene, CYP2B6 was most active isozyme to catalyse the formation of styrene oxide from styrene. Styrene 40-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 9253143-12 1997 At the higher concentration (1.8 mM) of styrene, CYP2B6 was most active isozyme to catalyse the formation of styrene oxide from styrene. styrene oxide 109-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 9253143-12 1997 At the higher concentration (1.8 mM) of styrene, CYP2B6 was most active isozyme to catalyse the formation of styrene oxide from styrene. Styrene 109-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 9253143-14 1997 These results suggest that CYP2E1 and CYP1A2/1 are the main isoforms responsible for the metabolism of toluene at low substrate concentrations in human liver microsomes, CYP2E1 at low styrene concentration, and CYP2C8 and CYP2B6 at high concentrations of toluene and styrene respectively. Toluene 103-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 222-228 9157990-4 1997 These inductions were associated with corresponding increases in immunoreactive CYP2B6, CYP2C8, CYP2C9, and CYP3A4, all previously shown to catalyze oxazaphosphorine activation. oxazaphosphorine 149-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 9113273-1 1997 Glycerol is widely used to stabilize cytochrome P-450 and prevent its transformation to cytochrome P-420. Glycerol 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-53 9155150-8 1997 Cancer cell lines indicated resistant to anti-cancer drugs, such as mitomycin C, doxorubicin, tamoxifen, cyclophosphamide and their derivatives, by a high activity of GST and a low activity of P450 in general. Doxorubicin 81-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 9155150-8 1997 Cancer cell lines indicated resistant to anti-cancer drugs, such as mitomycin C, doxorubicin, tamoxifen, cyclophosphamide and their derivatives, by a high activity of GST and a low activity of P450 in general. Cyclophosphamide 105-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 9241656-5 1997 Orphenadrine (ORP), a reported specific CYP2B6 inhibitor, was a less potent inhibitor of 7-HFC formation by microsomes from human liver than DDC or ANF. Orphenadrine 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 9241656-5 1997 Orphenadrine (ORP), a reported specific CYP2B6 inhibitor, was a less potent inhibitor of 7-HFC formation by microsomes from human liver than DDC or ANF. Orphenadrine 14-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 9241656-8 1997 Methoxychlor and S-mephenytoin inhibited expressed CYP2B6 but not CYP1A2 mediated 7-EFC O-deethylation. Methoxychlor 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 9241656-8 1997 Methoxychlor and S-mephenytoin inhibited expressed CYP2B6 but not CYP1A2 mediated 7-EFC O-deethylation. Mephenytoin 17-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 9237426-0 1997 Synthesis and in vitro evaluation of 3-(1-azolylmethyl)-1H-indoles and 3-(1-azolyl-1-phenylmethyl)-1H-indoles as inhibitors of P450 arom. 3-(1-azolylmethyl)-1h-indoles 37-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 9237426-0 1997 Synthesis and in vitro evaluation of 3-(1-azolylmethyl)-1H-indoles and 3-(1-azolyl-1-phenylmethyl)-1H-indoles as inhibitors of P450 arom. 3-(1-azolyl-1-phenylmethyl)-1h-indoles 71-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 9237426-1 1997 In the challenge to develop potent inhibitors of aromatase for reducing the levels of estrogens, we found that azolyl-substituted indoles inhibit aromatase activity, 3-(1-Azolylmethyl)-1H-indoles 9-15 and 3-(1-azolyl-1-phenylmethyl)-1H-indoles 22-25 were prepared, and tested on their ability to inhibit P450 arom. azolyl-substituted indoles 111-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 304-308 9179987-2 1997 The construction of three-dimensional models of CYP2B isozymes from rat (CYP2B1), rabbit (CYP2B4) and man (CYP2B6), based on a multiple sequence alignment with CYP102, a unique eukaryotic-like bacterial P450 (in terms of possessing an NADPH-dependent FAD- and FMN-containing oxidoreductase redox partner) of known crystal structure, is reported. NADP 235-240 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-53 9179987-2 1997 The construction of three-dimensional models of CYP2B isozymes from rat (CYP2B1), rabbit (CYP2B4) and man (CYP2B6), based on a multiple sequence alignment with CYP102, a unique eukaryotic-like bacterial P450 (in terms of possessing an NADPH-dependent FAD- and FMN-containing oxidoreductase redox partner) of known crystal structure, is reported. NADP 235-240 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 9179987-2 1997 The construction of three-dimensional models of CYP2B isozymes from rat (CYP2B1), rabbit (CYP2B4) and man (CYP2B6), based on a multiple sequence alignment with CYP102, a unique eukaryotic-like bacterial P450 (in terms of possessing an NADPH-dependent FAD- and FMN-containing oxidoreductase redox partner) of known crystal structure, is reported. Flavin-Adenine Dinucleotide 251-254 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-53 9179987-2 1997 The construction of three-dimensional models of CYP2B isozymes from rat (CYP2B1), rabbit (CYP2B4) and man (CYP2B6), based on a multiple sequence alignment with CYP102, a unique eukaryotic-like bacterial P450 (in terms of possessing an NADPH-dependent FAD- and FMN-containing oxidoreductase redox partner) of known crystal structure, is reported. Flavin-Adenine Dinucleotide 251-254 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 9113273-2 1997 The effect of glycerol on the content and activity of human cytochrome P-4502E1 (CYP2E1) in a HepG2 cell line that stably and constitutively expresses this P-450 was evaluated by immunoassays and oxidation of p-nitrophenol. Glycerol 14-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-76 9113273-2 1997 The effect of glycerol on the content and activity of human cytochrome P-4502E1 (CYP2E1) in a HepG2 cell line that stably and constitutively expresses this P-450 was evaluated by immunoassays and oxidation of p-nitrophenol. 4-nitrophenol 209-222 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-76 9107543-4 1997 The following catalytic markers of human P450 isozymes were used: ethoxyresorufin O-deethylase (P450s 1A1 and 1A2), diclofenac 4-hydroxylation (P4502C9), dextromethorphan O-demethylase (P4502D6), and testosterone 6 beta-hydroxylation (P4503A4). Diclofenac 116-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 9134230-6 1997 The potential significance of P450 inhibition by anaesthetics resides in the contribution of this enzyme family, in conjunction with that of cyclo-oxygenases and lipoxygenases, to the generation from arachidonic acid of lipid second messengers, the eicosanoids. Arachidonic Acid 200-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 9134230-6 1997 The potential significance of P450 inhibition by anaesthetics resides in the contribution of this enzyme family, in conjunction with that of cyclo-oxygenases and lipoxygenases, to the generation from arachidonic acid of lipid second messengers, the eicosanoids. Eicosanoids 249-260 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 9134230-8 1997 We have shown that P450 enzymes model the site of general anaesthesia in the tadpole with respect to (a) an absolute sensitivity to increasing chain-length series of flexible, straight chain primary and secondary alcohols and straight chain diols, (b) an absolute sensitivity to increasing molecular weight series of rigid cyclic alkanols and cyclic alkanemethanols, (c) the points of abrupt change and of reversal (cut-off) in the linear relationship between increasing anaesthetic potency with increasing carbon chain length, and (d) non-differentiation between secondary alkanol enantiomers. Alcohols 213-221 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 9134230-8 1997 We have shown that P450 enzymes model the site of general anaesthesia in the tadpole with respect to (a) an absolute sensitivity to increasing chain-length series of flexible, straight chain primary and secondary alcohols and straight chain diols, (b) an absolute sensitivity to increasing molecular weight series of rigid cyclic alkanols and cyclic alkanemethanols, (c) the points of abrupt change and of reversal (cut-off) in the linear relationship between increasing anaesthetic potency with increasing carbon chain length, and (d) non-differentiation between secondary alkanol enantiomers. diols 241-246 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 9134230-8 1997 We have shown that P450 enzymes model the site of general anaesthesia in the tadpole with respect to (a) an absolute sensitivity to increasing chain-length series of flexible, straight chain primary and secondary alcohols and straight chain diols, (b) an absolute sensitivity to increasing molecular weight series of rigid cyclic alkanols and cyclic alkanemethanols, (c) the points of abrupt change and of reversal (cut-off) in the linear relationship between increasing anaesthetic potency with increasing carbon chain length, and (d) non-differentiation between secondary alkanol enantiomers. cyclic alkanols 323-338 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 9134230-8 1997 We have shown that P450 enzymes model the site of general anaesthesia in the tadpole with respect to (a) an absolute sensitivity to increasing chain-length series of flexible, straight chain primary and secondary alcohols and straight chain diols, (b) an absolute sensitivity to increasing molecular weight series of rigid cyclic alkanols and cyclic alkanemethanols, (c) the points of abrupt change and of reversal (cut-off) in the linear relationship between increasing anaesthetic potency with increasing carbon chain length, and (d) non-differentiation between secondary alkanol enantiomers. alkanemethanols 350-365 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 9134230-8 1997 We have shown that P450 enzymes model the site of general anaesthesia in the tadpole with respect to (a) an absolute sensitivity to increasing chain-length series of flexible, straight chain primary and secondary alcohols and straight chain diols, (b) an absolute sensitivity to increasing molecular weight series of rigid cyclic alkanols and cyclic alkanemethanols, (c) the points of abrupt change and of reversal (cut-off) in the linear relationship between increasing anaesthetic potency with increasing carbon chain length, and (d) non-differentiation between secondary alkanol enantiomers. Carbon 507-513 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 9134230-8 1997 We have shown that P450 enzymes model the site of general anaesthesia in the tadpole with respect to (a) an absolute sensitivity to increasing chain-length series of flexible, straight chain primary and secondary alcohols and straight chain diols, (b) an absolute sensitivity to increasing molecular weight series of rigid cyclic alkanols and cyclic alkanemethanols, (c) the points of abrupt change and of reversal (cut-off) in the linear relationship between increasing anaesthetic potency with increasing carbon chain length, and (d) non-differentiation between secondary alkanol enantiomers. alkanol 330-337 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 9065730-6 1997 In addition, CYP2B6 and CYP2E1 catalyzed the formation of p-cresol (11-12% of total metabolites), and CYP1A2 catalyzed the formation of both o-(22%) and p-cresol (35%). 4-cresol 153-161 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 9172960-5 1997 In the preincubation protocol, orphenadrine decreased the CYP2B6 activity in cDNA-expressed cell microsomes to completion. Orphenadrine 31-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 9172960-6 1997 In human liver microsomes, orphenadrine strongly decreased the marker activities of CYP2B6, CYP2D6, as well as CYP2C9; and partially decreased the marker activities of CYP1A2, CYP2A6, CYP3A4, and CYP2C19. Orphenadrine 27-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 9065730-6 1997 In addition, CYP2B6 and CYP2E1 catalyzed the formation of p-cresol (11-12% of total metabolites), and CYP1A2 catalyzed the formation of both o-(22%) and p-cresol (35%). 4-cresol 58-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 9172960-0 1997 Orphenadrine and methimazole inhibit multiple cytochrome P450 enzymes in human liver microsomes. Orphenadrine 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 9172960-0 1997 Orphenadrine and methimazole inhibit multiple cytochrome P450 enzymes in human liver microsomes. Methimazole 17-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 9172960-1 1997 The specificities of orphenadrine and methimazole on eight human liver P450 enzyme activities were evaluated by studying the extent of inhibition at different concentrations in two protocols: competitive inhibition and preincubation. Orphenadrine 21-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 9172960-1 1997 The specificities of orphenadrine and methimazole on eight human liver P450 enzyme activities were evaluated by studying the extent of inhibition at different concentrations in two protocols: competitive inhibition and preincubation. Methimazole 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 9172960-2 1997 In the competitive inhibition protocol, orphenadrine decreased CYP2B6 marker activity up to 45-57% in human liver microsomes and up to 80-97% in cell microsomes containing cDNA-expressed CYP2B6. Orphenadrine 40-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 9172960-2 1997 In the competitive inhibition protocol, orphenadrine decreased CYP2B6 marker activity up to 45-57% in human liver microsomes and up to 80-97% in cell microsomes containing cDNA-expressed CYP2B6. Orphenadrine 40-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-193 9065730-9 1997 These results may explain, in part, the lower activity of CYP2B6, which has Phe at position 58 of the protein, for toluene ring oxidations than that of CYP2B1. Phenylalanine 76-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 9065730-9 1997 These results may explain, in part, the lower activity of CYP2B6, which has Phe at position 58 of the protein, for toluene ring oxidations than that of CYP2B1. Toluene 115-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-64 8960070-4 1997 The production rate of the main metabolites of colchicine"s 3-demethylcolchicine (3DMC) and 2-demethylcolchicine (2DMC), was linear in relation to incubation time, cytochrome (P450) content, and substrate concentration. Colchicine 47-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-180 9029055-5 1997 When radiolabeled PCP was used to inactivate the enzyme and the reaction mixture analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, radioactivity was found to be associated with P450, reductase, and catalase that had been added to protect against oxidative damage. Phencyclidine 18-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-213 8960070-4 1997 The production rate of the main metabolites of colchicine"s 3-demethylcolchicine (3DMC) and 2-demethylcolchicine (2DMC), was linear in relation to incubation time, cytochrome (P450) content, and substrate concentration. 3-desmethylcolchicine 60-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-180 8960070-4 1997 The production rate of the main metabolites of colchicine"s 3-demethylcolchicine (3DMC) and 2-demethylcolchicine (2DMC), was linear in relation to incubation time, cytochrome (P450) content, and substrate concentration. 3-desmethylcolchicine 82-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-180 8960070-4 1997 The production rate of the main metabolites of colchicine"s 3-demethylcolchicine (3DMC) and 2-demethylcolchicine (2DMC), was linear in relation to incubation time, cytochrome (P450) content, and substrate concentration. 2-Demethylcolchicine 92-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-180 8960070-4 1997 The production rate of the main metabolites of colchicine"s 3-demethylcolchicine (3DMC) and 2-demethylcolchicine (2DMC), was linear in relation to incubation time, cytochrome (P450) content, and substrate concentration. 2dmc 114-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-180 9332469-4 1997 CONCLUSIONS: Previous studies have attributed the production of the (S)-2-DCE-IFF and (S)-3-DCE-IFF metabolites to the activity of CYP2B6 and (R)-2-DCE-IFF and (R)-3-DCE-IFF to the activity of CYP3A4. (s)-2-dce-iff 68-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 9332469-4 1997 CONCLUSIONS: Previous studies have attributed the production of the (S)-2-DCE-IFF and (S)-3-DCE-IFF metabolites to the activity of CYP2B6 and (R)-2-DCE-IFF and (R)-3-DCE-IFF to the activity of CYP3A4. (s)-3-dce 86-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 9332469-4 1997 CONCLUSIONS: Previous studies have attributed the production of the (S)-2-DCE-IFF and (S)-3-DCE-IFF metabolites to the activity of CYP2B6 and (R)-2-DCE-IFF and (R)-3-DCE-IFF to the activity of CYP3A4. (r)-3-dce-iff 160-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 9332469-5 1997 The results suggest that phenytoin induced the activity of CYP2B6 to a greater extent than CYP3A4. Phenytoin 25-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 9046019-4 1997 Using a reconstituted system comprised of purified NADPH-P450 reductase, P450 and isolated microsomal lipid or pure L-alpha-phosphatidylcholine diarachidoyl, a mechanism has been proposed for the iron-independent microsomal lipid peroxidation and its prevention by ascorbic acid. l-alpha-phosphatidylcholine 116-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 9272409-9 1997 CONCLUSION: We conclude that for the non-response seen with maprotiline, P450 isozymes other than CYP2D6 or CYP1A2 are responsible. Maprotiline 60-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 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 2 subfamily B member 6 Homo sapiens 97-101 9046019-4 1997 Using a reconstituted system comprised of purified NADPH-P450 reductase, P450 and isolated microsomal lipid or pure L-alpha-phosphatidylcholine diarachidoyl, a mechanism has been proposed for the iron-independent microsomal lipid peroxidation and its prevention by ascorbic acid. l-alpha-phosphatidylcholine 116-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 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 2 subfamily B member 6 Homo sapiens 97-101 9046019-4 1997 Using a reconstituted system comprised of purified NADPH-P450 reductase, P450 and isolated microsomal lipid or pure L-alpha-phosphatidylcholine diarachidoyl, a mechanism has been proposed for the iron-independent microsomal lipid peroxidation and its prevention by ascorbic acid. diarachidoyl 144-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 9046019-4 1997 Using a reconstituted system comprised of purified NADPH-P450 reductase, P450 and isolated microsomal lipid or pure L-alpha-phosphatidylcholine diarachidoyl, a mechanism has been proposed for the iron-independent microsomal lipid peroxidation and its prevention by ascorbic acid. diarachidoyl 144-156 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 9046019-4 1997 Using a reconstituted system comprised of purified NADPH-P450 reductase, P450 and isolated microsomal lipid or pure L-alpha-phosphatidylcholine diarachidoyl, a mechanism has been proposed for the iron-independent microsomal lipid peroxidation and its prevention by ascorbic acid. Iron 196-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 9046019-4 1997 Using a reconstituted system comprised of purified NADPH-P450 reductase, P450 and isolated microsomal lipid or pure L-alpha-phosphatidylcholine diarachidoyl, a mechanism has been proposed for the iron-independent microsomal lipid peroxidation and its prevention by ascorbic acid. Iron 196-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 9046019-4 1997 Using a reconstituted system comprised of purified NADPH-P450 reductase, P450 and isolated microsomal lipid or pure L-alpha-phosphatidylcholine diarachidoyl, a mechanism has been proposed for the iron-independent microsomal lipid peroxidation and its prevention by ascorbic acid. Ascorbic Acid 265-278 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 9046019-4 1997 Using a reconstituted system comprised of purified NADPH-P450 reductase, P450 and isolated microsomal lipid or pure L-alpha-phosphatidylcholine diarachidoyl, a mechanism has been proposed for the iron-independent microsomal lipid peroxidation and its prevention by ascorbic acid. Ascorbic Acid 265-278 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 9046019-5 1997 It is proposed that the perferryl moiety P450 Fe3+.O2.- initiates lipid peroxidation by abstracting methylene hydrogen from polyunsaturated lipid to form lipid radical, which then combines with oxygen to produce the chain propagating peroxyl radical for subsequent formation of lipid peroxides. Oxygen 51-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 9046019-5 1997 It is proposed that the perferryl moiety P450 Fe3+.O2.- initiates lipid peroxidation by abstracting methylene hydrogen from polyunsaturated lipid to form lipid radical, which then combines with oxygen to produce the chain propagating peroxyl radical for subsequent formation of lipid peroxides. carbene 100-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 9046019-5 1997 It is proposed that the perferryl moiety P450 Fe3+.O2.- initiates lipid peroxidation by abstracting methylene hydrogen from polyunsaturated lipid to form lipid radical, which then combines with oxygen to produce the chain propagating peroxyl radical for subsequent formation of lipid peroxides. Hydrogen 110-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 9046019-5 1997 It is proposed that the perferryl moiety P450 Fe3+.O2.- initiates lipid peroxidation by abstracting methylene hydrogen from polyunsaturated lipid to form lipid radical, which then combines with oxygen to produce the chain propagating peroxyl radical for subsequent formation of lipid peroxides. polyunsaturated lipid 124-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 9046019-5 1997 It is proposed that the perferryl moiety P450 Fe3+.O2.- initiates lipid peroxidation by abstracting methylene hydrogen from polyunsaturated lipid to form lipid radical, which then combines with oxygen to produce the chain propagating peroxyl radical for subsequent formation of lipid peroxides. lipid radical 154-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 9046019-5 1997 It is proposed that the perferryl moiety P450 Fe3+.O2.- initiates lipid peroxidation by abstracting methylene hydrogen from polyunsaturated lipid to form lipid radical, which then combines with oxygen to produce the chain propagating peroxyl radical for subsequent formation of lipid peroxides. Oxygen 194-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 9046019-5 1997 It is proposed that the perferryl moiety P450 Fe3+.O2.- initiates lipid peroxidation by abstracting methylene hydrogen from polyunsaturated lipid to form lipid radical, which then combines with oxygen to produce the chain propagating peroxyl radical for subsequent formation of lipid peroxides. perhydroxyl radical 234-249 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 9046019-5 1997 It is proposed that the perferryl moiety P450 Fe3+.O2.- initiates lipid peroxidation by abstracting methylene hydrogen from polyunsaturated lipid to form lipid radical, which then combines with oxygen to produce the chain propagating peroxyl radical for subsequent formation of lipid peroxides. Lipid Peroxides 278-293 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 9046019-6 1997 Apparently, ascorbic acid prevents initiation of lipid peroxidation by interacting with P450 Fe3+.O2.-. Ascorbic Acid 12-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 9046019-6 1997 Apparently, ascorbic acid prevents initiation of lipid peroxidation by interacting with P450 Fe3+.O2.-. Oxygen 98-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 8954573-1 1996 The kinetics of CO binding to human cytochrome P450 1A1 was used to probe the interaction of polycyclic aromatic hydrocarbons (PAHs) with the membrane-bound P450 expressed in baculovirus-infected SF9 insect cells. Polycyclic Aromatic Hydrocarbons 93-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 9021169-1 1996 The metabolism of isoprene was investigated with microsomes derived from cell lines expressing human CYP1A1, CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2D6, CYP2E1, or CYP3A4. isoprene 18-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 8954573-1 1996 The kinetics of CO binding to human cytochrome P450 1A1 was used to probe the interaction of polycyclic aromatic hydrocarbons (PAHs) with the membrane-bound P450 expressed in baculovirus-infected SF9 insect cells. Polycyclic Aromatic Hydrocarbons 127-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 9021169-3 1996 CYP2E1 showed the highest rates of formation of the isoprene monoepoxides 3,4-epoxy-3-methyl-1-butene (EPOX-I) and 3,4-epoxy-2-methyl-1-butene (EPOX-II), followed by CYP2B6. isoprene monoepoxides 52-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 8954573-9 1996 In addition to PAHs, the interactions of P450 1A1 with 7-ethoxy- and 7-pentoxyresorufin were likewise examined for their effect on the CO binding kinetics. 7-ethoxy- and 7-pentoxyresorufin 55-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 8954573-11 1996 The markedly variable effects of these PAHs and resorufins on the CO binding kinetics indicate differential modes of interaction with the two target P450 1A1 species, resulting in differential modulation of their conformations. Polycyclic Aromatic Hydrocarbons 39-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-153 8954573-11 1996 The markedly variable effects of these PAHs and resorufins on the CO binding kinetics indicate differential modes of interaction with the two target P450 1A1 species, resulting in differential modulation of their conformations. resorufin 48-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-153 8989918-2 1996 In this study, we used eight human forms, four rodent forms, and one rabbit form of P450 expressed from recombinant vaccinia or baculovirus vectors to define their specificity for metabolizing DMBA. 9,10-Dimethyl-1,2-benzanthracene 193-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 8989918-3 1996 Of the eight human P450s, 1A1 was the most active (specific activity = 14.7 nmol/min/nmol of P450) in total metabolism of DMBA and showed approximately 6- to 33-fold more activity than other P450s, 2B6, 2C9, and 1A2 were also capable of metabolizing DMBA (2.0-2.5 nmol/min/nmol of P450), whereas 2C8, 2E1, 3A4, and 3A5 exhibited relatively low activities. 9,10-Dimethyl-1,2-benzanthracene 122-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 8989918-3 1996 Of the eight human P450s, 1A1 was the most active (specific activity = 14.7 nmol/min/nmol of P450) in total metabolism of DMBA and showed approximately 6- to 33-fold more activity than other P450s, 2B6, 2C9, and 1A2 were also capable of metabolizing DMBA (2.0-2.5 nmol/min/nmol of P450), whereas 2C8, 2E1, 3A4, and 3A5 exhibited relatively low activities. 9,10-Dimethyl-1,2-benzanthracene 122-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 8989918-3 1996 Of the eight human P450s, 1A1 was the most active (specific activity = 14.7 nmol/min/nmol of P450) in total metabolism of DMBA and showed approximately 6- to 33-fold more activity than other P450s, 2B6, 2C9, and 1A2 were also capable of metabolizing DMBA (2.0-2.5 nmol/min/nmol of P450), whereas 2C8, 2E1, 3A4, and 3A5 exhibited relatively low activities. 9,10-Dimethyl-1,2-benzanthracene 250-254 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 8989918-3 1996 Of the eight human P450s, 1A1 was the most active (specific activity = 14.7 nmol/min/nmol of P450) in total metabolism of DMBA and showed approximately 6- to 33-fold more activity than other P450s, 2B6, 2C9, and 1A2 were also capable of metabolizing DMBA (2.0-2.5 nmol/min/nmol of P450), whereas 2C8, 2E1, 3A4, and 3A5 exhibited relatively low activities. 9,10-Dimethyl-1,2-benzanthracene 250-254 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 8968067-11 1996 SKF525A, an isozyme non-selective P450 inhibitor, enhanced [3H]AFB1 binding to cellular DNA in unseparated cells, macrophages, and type II cells, suggesting that P450-mediated bioactivation of AFB1 is not a major pathway by which AFB1-8,9-epoxide is formed in human lung cells. Proadifen 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 8968067-11 1996 SKF525A, an isozyme non-selective P450 inhibitor, enhanced [3H]AFB1 binding to cellular DNA in unseparated cells, macrophages, and type II cells, suggesting that P450-mediated bioactivation of AFB1 is not a major pathway by which AFB1-8,9-epoxide is formed in human lung cells. Proadifen 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-166 8968067-5 1996 In whole lung microsomes, low levels of indomethacin inhibitable prostaglandin H synthase (PHS)-mediated [3H]AFB1-DNA binding and cytochrome P-450 (P450)-mediated [3H]AFB1-DNA binding were observed. Indomethacin 40-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-152 8968067-11 1996 SKF525A, an isozyme non-selective P450 inhibitor, enhanced [3H]AFB1 binding to cellular DNA in unseparated cells, macrophages, and type II cells, suggesting that P450-mediated bioactivation of AFB1 is not a major pathway by which AFB1-8,9-epoxide is formed in human lung cells. Tritium 60-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-166 8968067-5 1996 In whole lung microsomes, low levels of indomethacin inhibitable prostaglandin H synthase (PHS)-mediated [3H]AFB1-DNA binding and cytochrome P-450 (P450)-mediated [3H]AFB1-DNA binding were observed. Tritium 164-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-152 8968067-11 1996 SKF525A, an isozyme non-selective P450 inhibitor, enhanced [3H]AFB1 binding to cellular DNA in unseparated cells, macrophages, and type II cells, suggesting that P450-mediated bioactivation of AFB1 is not a major pathway by which AFB1-8,9-epoxide is formed in human lung cells. aflatoxin B1-2,3-oxide 230-246 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-166 8894508-1 1996 Omeprazole (OP) is a potent antiulcer drug that is metabolized by liver cytochrome P450 (P450) enzymes. Omeprazole 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-93 8948091-2 1996 We have examined the metabolism of diazepam by ten human cytochrome P450 forms (CYP1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4 and 3A5) expressed in HepG2 cells using a recombinant vaccinia virus system. Diazepam 35-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 8948091-4 1996 Among the P450 forms tested, diazepam was significantly demethylated by CYP2B6, 2C9, 2C19, 3A4 and 3A5, with 2C19 exhibiting the highest rate at concentrations < 0.1 mM, and hydroxylated only by the latter three enzymes, with 3A5 being the most active. Diazepam 29-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-14 8948091-4 1996 Among the P450 forms tested, diazepam was significantly demethylated by CYP2B6, 2C9, 2C19, 3A4 and 3A5, with 2C19 exhibiting the highest rate at concentrations < 0.1 mM, and hydroxylated only by the latter three enzymes, with 3A5 being the most active. Diazepam 29-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 8948091-13 1996 Our results suggest that in the human liver, the metabolism of diazepam to nordiazepam is mediated by CYP3A4, which has been reported as the most abundant P450 form in human liver as well as 2C19, which has been reported as a polymorphic enzyme. Diazepam 63-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-159 8948091-13 1996 Our results suggest that in the human liver, the metabolism of diazepam to nordiazepam is mediated by CYP3A4, which has been reported as the most abundant P450 form in human liver as well as 2C19, which has been reported as a polymorphic enzyme. Nordazepam 75-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 155-159 8900410-2 1996 Thromboxane synthase (TXAS), which catalyzes the biosynthesis of TXA, is a member of the cytochrome P450 superfamily. Thromboxane A2 22-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-104 8894508-1 1996 Omeprazole (OP) is a potent antiulcer drug that is metabolized by liver cytochrome P450 (P450) enzymes. Omeprazole 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-87 8969926-6 1996 Incubation of the purified enzyme with steroid in a reaction vessel containing a platinum electrode and a Ag/AgCl electrode couple poised at -650 mV, together with the electromotively active redox mediator, cobalt sepulchrate, results in the 17 alpha-hydroxylation of progesterone at rates as high as 25 nmoles of progesterone hydroxylated/min/nmole of P450. cobalt sepulchrate 207-225 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 353-357 8969926-6 1996 Incubation of the purified enzyme with steroid in a reaction vessel containing a platinum electrode and a Ag/AgCl electrode couple poised at -650 mV, together with the electromotively active redox mediator, cobalt sepulchrate, results in the 17 alpha-hydroxylation of progesterone at rates as high as 25 nmoles of progesterone hydroxylated/min/nmole of P450. Progesterone 268-280 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 353-357 8969926-6 1996 Incubation of the purified enzyme with steroid in a reaction vessel containing a platinum electrode and a Ag/AgCl electrode couple poised at -650 mV, together with the electromotively active redox mediator, cobalt sepulchrate, results in the 17 alpha-hydroxylation of progesterone at rates as high as 25 nmoles of progesterone hydroxylated/min/nmole of P450. Progesterone 314-326 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 353-357 8969926-8 1996 Similar experiments have been carried out for the generation of the 6 beta-hydroxylation product of testosterone (using a fusion protein containing human P450 3A4). Testosterone 100-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-158 8969926-9 1996 It is apparent that this method is applicable to many other P450 catalyzed reactions for the synthesis of large amounts of hydroxylated steroid metabolites. Steroids 136-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-64 8902262-4 1996 All drugs metabolized by P450 2D6 contain a basic nitrogen atom, and a flat hydrophobic region coplanar to the oxidation site which is either 5 or 7 A away from the basic nitrogen atom. Nitrogen 50-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 8902262-4 1996 All drugs metabolized by P450 2D6 contain a basic nitrogen atom, and a flat hydrophobic region coplanar to the oxidation site which is either 5 or 7 A away from the basic nitrogen atom. Nitrogen 171-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 8894508-9 1996 Recombinant human CYP3A4 formed 5OH-OP and OP-S with turnover numbers of 5.7 +/- 1.1 and 7.4 +/- 0.9 nmol/min/nmol of P450, respectively, and formed a minor unidentified metabolite. 5oh 32-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 8870687-2 1996 In this study, the oxidative bioactivation of 1,2-DBE to 2-bromoacetaldehyde (2-BA) was studied using heterologously expressed human cytochrome P450 (P450) isoenzymes and human liver microsomes. Ethylene Dibromide 46-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-155 8870687-2 1996 In this study, the oxidative bioactivation of 1,2-DBE to 2-bromoacetaldehyde (2-BA) was studied using heterologously expressed human cytochrome P450 (P450) isoenzymes and human liver microsomes. bromoacetaldehyde 57-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-155 8870687-2 1996 In this study, the oxidative bioactivation of 1,2-DBE to 2-bromoacetaldehyde (2-BA) was studied using heterologously expressed human cytochrome P450 (P450) isoenzymes and human liver microsomes. bromoacetaldehyde 78-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-155 8806785-0 1996 The interaction of NADPH-P450 reductase with P450: an electrochemical study of the role of the flavin mononucleotide-binding domain. Flavin Mononucleotide 95-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 8806763-0 1996 Kinetic analysis of the activation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone by heterologously expressed human P450 enzymes and the effect of P450-specific chemical inhibitors on this activation in human liver microsomes. 4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone 38-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-123 8806763-2 1996 The present study examined the kinetic parameters of NNK-derived keto alcohol (alpha-methyl hydroxylation), and keto aldehyde (alpha-methylene hydroxylation) formation catalyzed by human P450s heterologously expressed by either the baculovirus-insect cell expression system (P450s 2A6, 2D6, 2E1, and 3A4) or by stable expression in CHO cells (P450s 3A4 and 2D6) and human B-lymphoblastoid cells (P450 2D6). keto alcohol 65-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-191 8806763-2 1996 The present study examined the kinetic parameters of NNK-derived keto alcohol (alpha-methyl hydroxylation), and keto aldehyde (alpha-methylene hydroxylation) formation catalyzed by human P450s heterologously expressed by either the baculovirus-insect cell expression system (P450s 2A6, 2D6, 2E1, and 3A4) or by stable expression in CHO cells (P450s 3A4 and 2D6) and human B-lymphoblastoid cells (P450 2D6). keto aldehyde 112-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 187-191 8806763-4 1996 Human P450 2A6 showed the lowest KM (118 microM) for the formation of keto aldehyde. keto aldehyde 70-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-10 8806763-5 1996 A similar KM was observed for keto alcohol formation by expressed P450 2A6, but the kcat was lower than the value obtained for keto aldehyde formation. keto alcohol 30-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 8806763-5 1996 A similar KM was observed for keto alcohol formation by expressed P450 2A6, but the kcat was lower than the value obtained for keto aldehyde formation. keto aldehyde 127-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 8806763-8 1996 Expressed human P450 3A4 oxidized NNK to keto aldehyde also with a high KM. keto aldehyde 41-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 8806763-10 1996 A positive correlation coefficient of 0.74 was found between keto aldehyde formation and both coumarin 7-hydroxylation (P450 2A6) and 6 beta-testosterone hydroxylation (3A4) activity in characterized human liver microsomes. keto aldehyde 61-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-124 8806763-10 1996 A positive correlation coefficient of 0.74 was found between keto aldehyde formation and both coumarin 7-hydroxylation (P450 2A6) and 6 beta-testosterone hydroxylation (3A4) activity in characterized human liver microsomes. coumarin 94-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-124 8806763-11 1996 Keto alcohol formation showed a significant correlation with ethoxyresorufin O-dealkylation (P450 1A2) in human liver microsomes. keto alcohol 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 8806763-11 1996 Keto alcohol formation showed a significant correlation with ethoxyresorufin O-dealkylation (P450 1A2) in human liver microsomes. ethoxyresorufin 61-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 8806763-12 1996 Both coumarin and troleandomycin, specific inhibitors of P450 2A6 and 3A4, respectively, inhibited the formation of keto aldehyde, but inhibited the formation of keto alcohol only slightly in human liver microsomes. coumarin 5-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8806763-12 1996 Both coumarin and troleandomycin, specific inhibitors of P450 2A6 and 3A4, respectively, inhibited the formation of keto aldehyde, but inhibited the formation of keto alcohol only slightly in human liver microsomes. Troleandomycin 18-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8806763-12 1996 Both coumarin and troleandomycin, specific inhibitors of P450 2A6 and 3A4, respectively, inhibited the formation of keto aldehyde, but inhibited the formation of keto alcohol only slightly in human liver microsomes. keto aldehyde 116-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8806763-12 1996 Both coumarin and troleandomycin, specific inhibitors of P450 2A6 and 3A4, respectively, inhibited the formation of keto aldehyde, but inhibited the formation of keto alcohol only slightly in human liver microsomes. keto alcohol 162-174 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8806763-13 1996 Both furafylline, a P450 1A2 inhibitor, and N-nitrosodimethylamine, a P450 2E1 substrate, inhibited the formation of keto alcohol but not keto aldehyde in human liver microsomes. furafylline 5-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 8806763-13 1996 Both furafylline, a P450 1A2 inhibitor, and N-nitrosodimethylamine, a P450 2E1 substrate, inhibited the formation of keto alcohol but not keto aldehyde in human liver microsomes. Dimethylnitrosamine 44-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 8806763-13 1996 Both furafylline, a P450 1A2 inhibitor, and N-nitrosodimethylamine, a P450 2E1 substrate, inhibited the formation of keto alcohol but not keto aldehyde in human liver microsomes. keto alcohol 117-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 8806763-13 1996 Both furafylline, a P450 1A2 inhibitor, and N-nitrosodimethylamine, a P450 2E1 substrate, inhibited the formation of keto alcohol but not keto aldehyde in human liver microsomes. keto alcohol 117-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 8806763-14 1996 Quinidine, a specific inhibitor of P450 2D6, was not an effective inhibitor of NNK metabolism. Quinidine 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 8886603-9 1996 In contrast, orphenadrine, an inhibitor of CYP2B forms, produced a 51 +/- 4% decrease in S-mephenytoin N-demethylase activity in human liver microsomes and a 45% decrease in recombinant microsomes expressing CYP2B6. Orphenadrine 13-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 208-214 8886603-12 1996 Taken together, these studies indicate that the N-demethylation of S-mephenytoin by human liver microsomes is catalyzed primarily by CYP2B6. Nitrogen 48-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 8886603-12 1996 Taken together, these studies indicate that the N-demethylation of S-mephenytoin by human liver microsomes is catalyzed primarily by CYP2B6. Mephenytoin 67-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-139 8806785-0 1996 The interaction of NADPH-P450 reductase with P450: an electrochemical study of the role of the flavin mononucleotide-binding domain. Flavin Mononucleotide 95-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 45-49 8806785-1 1996 The electrochemically reduced mediator cobalt sepulchrate requires the presence of a flavoprotein for the rapid transfer of electrons to cytochrome P450. cobalt sepulchrate 39-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 148-152 8794884-0 1996 Role of heme in cytochrome P450 transcription and function in mice treated with lead acetate. Heme 8-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 8794884-0 1996 Role of heme in cytochrome P450 transcription and function in mice treated with lead acetate. lead acetate 80-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 8794884-2 1996 The hypothetical explanations have been either a decreased supply of heme for saturation of apo-P450 or a requirement of heme for P450 gene transcription. Heme 69-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-100 8794884-2 1996 The hypothetical explanations have been either a decreased supply of heme for saturation of apo-P450 or a requirement of heme for P450 gene transcription. Heme 121-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-134 8794884-5 1996 P450-dependent activities (7-ethoxycoumarin O-deethylation and O-dealkylation of alkoxyresorufins) decreased progressively after lead injection to 56-69% of control levels within 20 hr. 7-ethoxycoumarin 27-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 8794884-10 1996 We conclude that the decrease of P450 in lead poisoning is a consequence of two different mechanisms: (a) a mechanism unrelated to heme, in which lead decreases P450 transcription; and (b) a mechanism dependent on heme, in which lead inhibits heme synthesis, and this results in a decreased heme saturation of P450 and/or apo-P450 content. Heme 131-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 8794884-10 1996 We conclude that the decrease of P450 in lead poisoning is a consequence of two different mechanisms: (a) a mechanism unrelated to heme, in which lead decreases P450 transcription; and (b) a mechanism dependent on heme, in which lead inhibits heme synthesis, and this results in a decreased heme saturation of P450 and/or apo-P450 content. Heme 214-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 8794884-10 1996 We conclude that the decrease of P450 in lead poisoning is a consequence of two different mechanisms: (a) a mechanism unrelated to heme, in which lead decreases P450 transcription; and (b) a mechanism dependent on heme, in which lead inhibits heme synthesis, and this results in a decreased heme saturation of P450 and/or apo-P450 content. Heme 214-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 8794884-10 1996 We conclude that the decrease of P450 in lead poisoning is a consequence of two different mechanisms: (a) a mechanism unrelated to heme, in which lead decreases P450 transcription; and (b) a mechanism dependent on heme, in which lead inhibits heme synthesis, and this results in a decreased heme saturation of P450 and/or apo-P450 content. Heme 214-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-37 8812189-10 1996 Lindane shares the criteria of the second and third groups and seems to induce both CYP1A and CYP2B activities. Hexachlorocyclohexane 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-99 8819299-4 1996 Coumarin, 7-ethoxyresorufin, 7-benzyloxyresorufin, tolbutamide, aniline and diazepam were form-selective substrates towards CYP2A6, the CYP1A subfamily, CYP2B6, the CYP2C subfamily, CYP2E1 and the CYP3A subfamily respectively. coumarin 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 8829341-3 1996 In humans, fluoroquinolones hinder the metabolism of other clinically important drugs through inhibition of hepatic cytochrome P-450"s (P450). Fluoroquinolones 11-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-134 8829341-3 1996 In humans, fluoroquinolones hinder the metabolism of other clinically important drugs through inhibition of hepatic cytochrome P-450"s (P450). Fluoroquinolones 11-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 8764331-7 1996 Among the recombinant human CYP isoforms, CYP2D6, 2B6, 3A4 and 1A2 catalyzed the 8-hydroxylation, and CYP1A2 and 3A4 were involved exclusively in the N-oxidation, whereas CYP2B6, 2C19, 1A2, 3A4 and 2D6 showed a catalytic activity for the N-demethylation, for either or both of MS enantiomers. Nitrogen 150-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 8819299-4 1996 Coumarin, 7-ethoxyresorufin, 7-benzyloxyresorufin, tolbutamide, aniline and diazepam were form-selective substrates towards CYP2A6, the CYP1A subfamily, CYP2B6, the CYP2C subfamily, CYP2E1 and the CYP3A subfamily respectively. ethoxyresorufin 10-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 8819299-4 1996 Coumarin, 7-ethoxyresorufin, 7-benzyloxyresorufin, tolbutamide, aniline and diazepam were form-selective substrates towards CYP2A6, the CYP1A subfamily, CYP2B6, the CYP2C subfamily, CYP2E1 and the CYP3A subfamily respectively. benzyloxyresorufin 29-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 8819299-4 1996 Coumarin, 7-ethoxyresorufin, 7-benzyloxyresorufin, tolbutamide, aniline and diazepam were form-selective substrates towards CYP2A6, the CYP1A subfamily, CYP2B6, the CYP2C subfamily, CYP2E1 and the CYP3A subfamily respectively. Tolbutamide 51-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 8819299-4 1996 Coumarin, 7-ethoxyresorufin, 7-benzyloxyresorufin, tolbutamide, aniline and diazepam were form-selective substrates towards CYP2A6, the CYP1A subfamily, CYP2B6, the CYP2C subfamily, CYP2E1 and the CYP3A subfamily respectively. aniline 64-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 8819299-4 1996 Coumarin, 7-ethoxyresorufin, 7-benzyloxyresorufin, tolbutamide, aniline and diazepam were form-selective substrates towards CYP2A6, the CYP1A subfamily, CYP2B6, the CYP2C subfamily, CYP2E1 and the CYP3A subfamily respectively. Diazepam 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 8819299-7 1996 SKF-525A inhibited > 40% of the metabolic activity of all substrates tested, and the inhibitory effects differed among P450 forms. Proadifen 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-126 8809764-4 1996 Much research is focussed on mammalian P-450s, with their roles in such processes as steroid transformations and the metabolism of carcinogens and other xenobiotics. Steroids 85-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 8651689-4 1996 After large scale labeling in the absence and in the presence of alpha-naphthoflavone, P-450 was digested with 1-p-tosyl-amino-2-phenylethyl chloromethyl ketone-treated trypsin and the resultant peptide fragments were separated with HPLC on a reverse-phase column. alpha-naphthoflavone 65-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-92 8651689-4 1996 After large scale labeling in the absence and in the presence of alpha-naphthoflavone, P-450 was digested with 1-p-tosyl-amino-2-phenylethyl chloromethyl ketone-treated trypsin and the resultant peptide fragments were separated with HPLC on a reverse-phase column. 1-p-tosyl-amino-2-phenylethyl chloromethyl ketone 111-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-92 8647857-1 1996 Human P450 2C19 is selective for 4"-hydroxylation of S-mephenytoin and 5-hydroxylation of omeprazole, while the structurally homologous P450 2C9 has low activity toward these substrates. Mephenytoin 53-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-10 8647857-1 1996 Human P450 2C19 is selective for 4"-hydroxylation of S-mephenytoin and 5-hydroxylation of omeprazole, while the structurally homologous P450 2C9 has low activity toward these substrates. Mephenytoin 53-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 8647857-1 1996 Human P450 2C19 is selective for 4"-hydroxylation of S-mephenytoin and 5-hydroxylation of omeprazole, while the structurally homologous P450 2C9 has low activity toward these substrates. Omeprazole 90-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-10 8647857-3 1996 The 339 NH2-terminal amino acid residues (SRS-1-SRS-4) and amino acids 160-383 (SRS-2-SRS-5) of P450 2C19 conferred omeprazole 5-hydroxylase activity to P450 2C9. Omeprazole 116-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-100 8647857-3 1996 The 339 NH2-terminal amino acid residues (SRS-1-SRS-4) and amino acids 160-383 (SRS-2-SRS-5) of P450 2C19 conferred omeprazole 5-hydroxylase activity to P450 2C9. Omeprazole 116-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-157 8647857-7 1996 A combination of two mutations, Ile99 --> His and Ser200 --> Pro, converted P450 2C9 to an enzyme with a turnover number of omeprazole 5-hyrdroxylation, which resembled that of P450 /c19. Histidine 45-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 8647857-7 1996 A combination of two mutations, Ile99 --> His and Ser200 --> Pro, converted P450 2C9 to an enzyme with a turnover number of omeprazole 5-hyrdroxylation, which resembled that of P450 /c19. Histidine 45-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 8647857-7 1996 A combination of two mutations, Ile99 --> His and Ser200 --> Pro, converted P450 2C9 to an enzyme with a turnover number of omeprazole 5-hyrdroxylation, which resembled that of P450 /c19. Proline 67-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 8647857-7 1996 A combination of two mutations, Ile99 --> His and Ser200 --> Pro, converted P450 2C9 to an enzyme with a turnover number of omeprazole 5-hyrdroxylation, which resembled that of P450 /c19. Proline 67-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 8647857-7 1996 A combination of two mutations, Ile99 --> His and Ser200 --> Pro, converted P450 2C9 to an enzyme with a turnover number of omeprazole 5-hyrdroxylation, which resembled that of P450 /c19. Omeprazole 130-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 8647857-7 1996 A combination of two mutations, Ile99 --> His and Ser200 --> Pro, converted P450 2C9 to an enzyme with a turnover number of omeprazole 5-hyrdroxylation, which resembled that of P450 /c19. Omeprazole 130-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 8647857-11 1996 Our results thus indicate that amino acids 99, 220, and 221 are key residues that determine the specificity of P450 2C19 for omeprazole. Omeprazole 125-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-115 8723730-8 1996 These results suggest that in human fetal livers at least two P450 enzymes, a form of P450 that is immunoreactive P4501A1 and P4503A7, are actually expressed and these enzymes are suggested as being involved in the activation of the (+)- and (-)-enantiomers of 7,8-dihydroxy-7,8-dihydrobenzo[a]pyrene and the carcinogenic mycotoxins, respectively. benzo(a)pyrene 7,8-dihydrodiol 261-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-66 8723730-8 1996 These results suggest that in human fetal livers at least two P450 enzymes, a form of P450 that is immunoreactive P4501A1 and P4503A7, are actually expressed and these enzymes are suggested as being involved in the activation of the (+)- and (-)-enantiomers of 7,8-dihydroxy-7,8-dihydrobenzo[a]pyrene and the carcinogenic mycotoxins, respectively. benzo(a)pyrene 7,8-dihydrodiol 261-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-90 8771373-5 1996 Both AF and BP are substrates for the P-450 and flavin-containing monooxygenase enzyme system, which can be induced with beta-naphthoflavone (beta NF). Benzo(a)pyrene 12-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-79 8771373-5 1996 Both AF and BP are substrates for the P-450 and flavin-containing monooxygenase enzyme system, which can be induced with beta-naphthoflavone (beta NF). beta-Naphthoflavone 121-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-79 8771373-5 1996 Both AF and BP are substrates for the P-450 and flavin-containing monooxygenase enzyme system, which can be induced with beta-naphthoflavone (beta NF). beta-Naphthoflavone 142-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-79 8640822-0 1996 Sensitization of human breast cancer cells to cyclophosphamide and ifosfamide by transfer of a liver cytochrome P450 gene. Cyclophosphamide 46-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 8627562-0 1996 Identification of the human P450 enzymes involved in lansoprazole metabolism. Lansoprazole 53-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-32 8627562-1 1996 The aim of this study was to identify which human P450 enzymes are involved in the metabolism of lansoprazole. Lansoprazole 97-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 8619853-3 1996 Using purified cDNA-expressed CYP3A4 incorporated into membranous vesicles made from microsomal phospholipids, rates of nifedipine and testosterone oxidation of about 60 nmol/nmol P450/min were achieved, whereas similar reconstitution into dilauroyl-phosphatidylcholine micelles was unsuccessful. Phospholipids 96-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 180-188 8619853-3 1996 Using purified cDNA-expressed CYP3A4 incorporated into membranous vesicles made from microsomal phospholipids, rates of nifedipine and testosterone oxidation of about 60 nmol/nmol P450/min were achieved, whereas similar reconstitution into dilauroyl-phosphatidylcholine micelles was unsuccessful. Nifedipine 120-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 180-188 8619853-3 1996 Using purified cDNA-expressed CYP3A4 incorporated into membranous vesicles made from microsomal phospholipids, rates of nifedipine and testosterone oxidation of about 60 nmol/nmol P450/min were achieved, whereas similar reconstitution into dilauroyl-phosphatidylcholine micelles was unsuccessful. Testosterone 135-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 180-188 8640822-1 1996 The cancer chemotherapeutic agent cyclophosphamide (CPA) and its isomer ifosfamide (IFA) are alkylating agent prodrugs that require metabolism by liver cytochrome P450 (P450) enzymes for antitumor activity. Cyclophosphamide 52-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-167 8640822-1 1996 The cancer chemotherapeutic agent cyclophosphamide (CPA) and its isomer ifosfamide (IFA) are alkylating agent prodrugs that require metabolism by liver cytochrome P450 (P450) enzymes for antitumor activity. Cyclophosphamide 52-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 8640822-1 1996 The cancer chemotherapeutic agent cyclophosphamide (CPA) and its isomer ifosfamide (IFA) are alkylating agent prodrugs that require metabolism by liver cytochrome P450 (P450) enzymes for antitumor activity. Ifosfamide 72-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-167 8866826-6 1996 Human CYP2B6 and rat CYP2B1 had high lidocaine N-deethylation activity. lidocaine n 37-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 8625481-4 1996 32P-Postlabeling assays showed that MCL-5 cells (genetically engineered to express five human cytochromes P450 and microsomal epoxide hydrolase) formed characteristic adduct spots with benz[a]pyrene, benzo[g]chrysene, 7,12-dimethylbenz[a]anthracene, benzidine, sterigmatocystin and 3-methylcholanthrene, whereas CCRF cells did not. Phosphorus-32 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-143 8625481-4 1996 32P-Postlabeling assays showed that MCL-5 cells (genetically engineered to express five human cytochromes P450 and microsomal epoxide hydrolase) formed characteristic adduct spots with benz[a]pyrene, benzo[g]chrysene, 7,12-dimethylbenz[a]anthracene, benzidine, sterigmatocystin and 3-methylcholanthrene, whereas CCRF cells did not. benzylideneacetone 185-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-143 8625481-4 1996 32P-Postlabeling assays showed that MCL-5 cells (genetically engineered to express five human cytochromes P450 and microsomal epoxide hydrolase) formed characteristic adduct spots with benz[a]pyrene, benzo[g]chrysene, 7,12-dimethylbenz[a]anthracene, benzidine, sterigmatocystin and 3-methylcholanthrene, whereas CCRF cells did not. pyrene 192-198 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-143 8625481-4 1996 32P-Postlabeling assays showed that MCL-5 cells (genetically engineered to express five human cytochromes P450 and microsomal epoxide hydrolase) formed characteristic adduct spots with benz[a]pyrene, benzo[g]chrysene, 7,12-dimethylbenz[a]anthracene, benzidine, sterigmatocystin and 3-methylcholanthrene, whereas CCRF cells did not. benzo(g)chrysene 200-216 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-143 8625481-4 1996 32P-Postlabeling assays showed that MCL-5 cells (genetically engineered to express five human cytochromes P450 and microsomal epoxide hydrolase) formed characteristic adduct spots with benz[a]pyrene, benzo[g]chrysene, 7,12-dimethylbenz[a]anthracene, benzidine, sterigmatocystin and 3-methylcholanthrene, whereas CCRF cells did not. 7,12-dimethylbenz 218-235 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-143 8625481-4 1996 32P-Postlabeling assays showed that MCL-5 cells (genetically engineered to express five human cytochromes P450 and microsomal epoxide hydrolase) formed characteristic adduct spots with benz[a]pyrene, benzo[g]chrysene, 7,12-dimethylbenz[a]anthracene, benzidine, sterigmatocystin and 3-methylcholanthrene, whereas CCRF cells did not. anthracene 238-248 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-143 8625481-4 1996 32P-Postlabeling assays showed that MCL-5 cells (genetically engineered to express five human cytochromes P450 and microsomal epoxide hydrolase) formed characteristic adduct spots with benz[a]pyrene, benzo[g]chrysene, 7,12-dimethylbenz[a]anthracene, benzidine, sterigmatocystin and 3-methylcholanthrene, whereas CCRF cells did not. benzidine 250-259 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-143 8625481-4 1996 32P-Postlabeling assays showed that MCL-5 cells (genetically engineered to express five human cytochromes P450 and microsomal epoxide hydrolase) formed characteristic adduct spots with benz[a]pyrene, benzo[g]chrysene, 7,12-dimethylbenz[a]anthracene, benzidine, sterigmatocystin and 3-methylcholanthrene, whereas CCRF cells did not. Sterigmatocystin 261-277 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-143 8625481-4 1996 32P-Postlabeling assays showed that MCL-5 cells (genetically engineered to express five human cytochromes P450 and microsomal epoxide hydrolase) formed characteristic adduct spots with benz[a]pyrene, benzo[g]chrysene, 7,12-dimethylbenz[a]anthracene, benzidine, sterigmatocystin and 3-methylcholanthrene, whereas CCRF cells did not. Methylcholanthrene 282-302 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-143 8640822-1 1996 The cancer chemotherapeutic agent cyclophosphamide (CPA) and its isomer ifosfamide (IFA) are alkylating agent prodrugs that require metabolism by liver cytochrome P450 (P450) enzymes for antitumor activity. Ifosfamide 72-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 8640822-1 1996 The cancer chemotherapeutic agent cyclophosphamide (CPA) and its isomer ifosfamide (IFA) are alkylating agent prodrugs that require metabolism by liver cytochrome P450 (P450) enzymes for antitumor activity. Ifosfamide 84-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-167 8640822-1 1996 The cancer chemotherapeutic agent cyclophosphamide (CPA) and its isomer ifosfamide (IFA) are alkylating agent prodrugs that require metabolism by liver cytochrome P450 (P450) enzymes for antitumor activity. Ifosfamide 84-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 8640822-3 1996 Transfer of a liver cytochrome P450 gene, CYP2B1, into human breast MCF-7 cancer cells is presently shown to greatly sensitize these cells to oxazaphosphorine toxicity as a consequence of the acquired capacity for intratumoral CPA and IFA activation. oxazaphosphorine 142-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 8640822-3 1996 Transfer of a liver cytochrome P450 gene, CYP2B1, into human breast MCF-7 cancer cells is presently shown to greatly sensitize these cells to oxazaphosphorine toxicity as a consequence of the acquired capacity for intratumoral CPA and IFA activation. Cyclophosphamide 227-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-35 8640822-0 1996 Sensitization of human breast cancer cells to cyclophosphamide and ifosfamide by transfer of a liver cytochrome P450 gene. Ifosfamide 67-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 8640822-1 1996 The cancer chemotherapeutic agent cyclophosphamide (CPA) and its isomer ifosfamide (IFA) are alkylating agent prodrugs that require metabolism by liver cytochrome P450 (P450) enzymes for antitumor activity. Cyclophosphamide 34-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-167 8640822-1 1996 The cancer chemotherapeutic agent cyclophosphamide (CPA) and its isomer ifosfamide (IFA) are alkylating agent prodrugs that require metabolism by liver cytochrome P450 (P450) enzymes for antitumor activity. Cyclophosphamide 34-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-173 9010600-2 1996 Cytochromes P450 have been implicated in transforming these compounds to more reactive species, under anaerobic conditions, through reduction at the heme. Heme 149-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-16 8621735-4 1996 The I172N apoprotein was synthesized at the normal rate, but its heme-bound P450 form was present at a much lower level. Heme 65-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 8573129-1 1996 Linoleic acid, the predominant polyunsaturated fatty acid in the diet, can be metabolized by cyclooxygenase, lipoxygenase and P450 enzymes. Linoleic Acid 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 8573129-1 1996 Linoleic acid, the predominant polyunsaturated fatty acid in the diet, can be metabolized by cyclooxygenase, lipoxygenase and P450 enzymes. Fatty Acids, Unsaturated 31-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 8632407-10 1996 P450 17: K(m)progesterone = 7 microM, K(i)16 = 9 microM, K(i)20 = 80 nM). Progesterone 13-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 8737761-1 1996 OBJECTIVE: The affinity of (+)-, (-)- and (+/-)- fluvastatin, a new synthetic HMG-CoA reductase inhibitor developed as a racemate, for specific human P450 monooxygenases in liver microsomes was compared with that of the pharmacologically active acidic forms of lovastatin, pravastatin and simvastatin. Simvastatin 289-300 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 8737761-3 1996 RESULTS: Lovastatin acid, pravastatin and simvastatin acid displayed moderate affinity for all three P450 isozymes (estimated Ki > 50 micromol.1(-1)). mevinolinic acid 9-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 8737761-1 1996 OBJECTIVE: The affinity of (+)-, (-)- and (+/-)- fluvastatin, a new synthetic HMG-CoA reductase inhibitor developed as a racemate, for specific human P450 monooxygenases in liver microsomes was compared with that of the pharmacologically active acidic forms of lovastatin, pravastatin and simvastatin. (+)-, (-)- and (+/-)- fluvastatin 27-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 8737761-1 1996 OBJECTIVE: The affinity of (+)-, (-)- and (+/-)- fluvastatin, a new synthetic HMG-CoA reductase inhibitor developed as a racemate, for specific human P450 monooxygenases in liver microsomes was compared with that of the pharmacologically active acidic forms of lovastatin, pravastatin and simvastatin. Lovastatin 261-271 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 8737761-3 1996 RESULTS: Lovastatin acid, pravastatin and simvastatin acid displayed moderate affinity for all three P450 isozymes (estimated Ki > 50 micromol.1(-1)). Pravastatin 26-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 8737761-1 1996 OBJECTIVE: The affinity of (+)-, (-)- and (+/-)- fluvastatin, a new synthetic HMG-CoA reductase inhibitor developed as a racemate, for specific human P450 monooxygenases in liver microsomes was compared with that of the pharmacologically active acidic forms of lovastatin, pravastatin and simvastatin. Pravastatin 273-284 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 8737761-3 1996 RESULTS: Lovastatin acid, pravastatin and simvastatin acid displayed moderate affinity for all three P450 isozymes (estimated Ki > 50 micromol.1(-1)). simvastatin acid 42-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 8558432-0 1996 Metabolism of 3-methylindole by vaccinia-expressed P450 enzymes: correlation of 3-methyleneindolenine formation and protein-binding. Skatole 14-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 8558432-0 1996 Metabolism of 3-methylindole by vaccinia-expressed P450 enzymes: correlation of 3-methyleneindolenine formation and protein-binding. 3-methyleneindolenine 80-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 8558432-1 1996 The toxicity of 3-methylindole (3 MI), a selective pneumotoxin, is dependent upon cytochrome P450-mediated bioactivation 3. Skatole 16-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 8558432-1 1996 The toxicity of 3-methylindole (3 MI), a selective pneumotoxin, is dependent upon cytochrome P450-mediated bioactivation 3. 2-methyl-4-isothiazolin-3-one 34-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 8558432-9 1996 The toxicity of 3 MI is believed to be due to covalent binding of a P450-generated intermediate to critical pulmonary proteins. 2-methyl-4-isothiazolin-3-one 18-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 8558432-11 1996 This study showed that the methylene imine electrophile is produced by only a few P450 enzymes and is the metabolite responsible for the covalent binding and presumably, the toxicity of 3 MI. methyleneimine 27-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 8558432-11 1996 This study showed that the methylene imine electrophile is produced by only a few P450 enzymes and is the metabolite responsible for the covalent binding and presumably, the toxicity of 3 MI. 2-methyl-4-isothiazolin-3-one 188-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 8839282-4 1996 Other important hepatocellular processes involving bile acids include active uptake by the basolateral membrane, intracellular transport, P-450-mediated conjugations and hydroxylations, and canalicular secretion. Bile Acids and Salts 51-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-143 8571359-2 1995 Many xenobiotics including pesticides, nitrosamines, polycyclic aromatic hydrocarbons and halogenated hydrocarbons, require bioactivation by P450 enzymes to elicit toxicity. Nitrosamines 39-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 8571359-2 1995 Many xenobiotics including pesticides, nitrosamines, polycyclic aromatic hydrocarbons and halogenated hydrocarbons, require bioactivation by P450 enzymes to elicit toxicity. Polycyclic Aromatic Hydrocarbons 53-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 8571359-2 1995 Many xenobiotics including pesticides, nitrosamines, polycyclic aromatic hydrocarbons and halogenated hydrocarbons, require bioactivation by P450 enzymes to elicit toxicity. Hydrocarbons 73-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 141-145 7585636-0 1995 Activation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in human lung microsomes by cytochromes P450, lipoxygenase, and hydroperoxides. 4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone 14-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-125 7585636-0 1995 Activation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in human lung microsomes by cytochromes P450, lipoxygenase, and hydroperoxides. 4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone 62-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-125 8654205-5 1995 The turnover rates (min-1) of DETC-Me sulfoxidation by the cDNA-expressed cytochrome P450 enzymes ranked as follows: CYP3A4 > CYP2A6 = CYP2C9 > CYP1A2 > CYP2B6 = CYP2E1 > CYP1A1 > CYP2D6. detc-me 30-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 8552139-0 1995 DNA adducts and P450 induction in human, rat and avian liver cells after exposure to polychlorobiphenyls. Polychlorinated Biphenyls 85-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 8703653-4 1995 The findings suggest that some SSRIs and their metabolites with a low Ki value (e.g., fluoxetine, norfluoxetine) may reduce the clearance of drugs metabolized by this isoform of P450, thereby resulting in a possible drug-drug interaction, when administered simultaneously. Fluoxetine 86-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 8703653-4 1995 The findings suggest that some SSRIs and their metabolites with a low Ki value (e.g., fluoxetine, norfluoxetine) may reduce the clearance of drugs metabolized by this isoform of P450, thereby resulting in a possible drug-drug interaction, when administered simultaneously. norfluoxetine 98-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-182 8578771-11 1995 The effects of the cytochrome P450 inhibitors, ketoconazole and metyrapone, indicated that P450 played a role in the toxicity of CsA but not tamoxifen. Ketoconazole 47-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 8578771-11 1995 The effects of the cytochrome P450 inhibitors, ketoconazole and metyrapone, indicated that P450 played a role in the toxicity of CsA but not tamoxifen. Metyrapone 64-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 8578771-11 1995 The effects of the cytochrome P450 inhibitors, ketoconazole and metyrapone, indicated that P450 played a role in the toxicity of CsA but not tamoxifen. Metyrapone 64-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 8578771-11 1995 The effects of the cytochrome P450 inhibitors, ketoconazole and metyrapone, indicated that P450 played a role in the toxicity of CsA but not tamoxifen. Cyclosporine 129-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 8578771-11 1995 The effects of the cytochrome P450 inhibitors, ketoconazole and metyrapone, indicated that P450 played a role in the toxicity of CsA but not tamoxifen. Cyclosporine 129-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-95 8597120-4 1995 The brain microsomal and mitochondrial P450 systems are capable of metabolizing a variety of xenobiotics, while the brain FMO efficiently metabolizes a variety of psychoactive drugs to their respective N-oxides. n-oxides 202-210 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 7626059-0 1995 Metabolism of highly persistent PCB congener, 2,4,5,2",4",5"-hexachlorobiphenyl, by human CYP2B6. 2,4,5,2',4',5'-hexachlorobiphenyl 46-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 8579473-6 1995 The specific biosynthetic aromatase cytochrome P-450 activity contained in the tissue system can synthesize [14C]estradiol-17 beta from [4-14C]testosterone. Carbon-14 109-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-52 8579473-6 1995 The specific biosynthetic aromatase cytochrome P-450 activity contained in the tissue system can synthesize [14C]estradiol-17 beta from [4-14C]testosterone. Estradiol 113-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-52 8579473-6 1995 The specific biosynthetic aromatase cytochrome P-450 activity contained in the tissue system can synthesize [14C]estradiol-17 beta from [4-14C]testosterone. Testosterone 143-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-52 7626059-1 1995 Metabolism of 2,4,5,2",4",5"-hexachlorobiphenyl was studied with cDNA-expressed human P450 2B isoform, CYP2B6. 2,4,5,2',4',5'-hexachlorobiphenyl 14-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 7625841-9 1995 After the dissociation of the oligomers by 0.2% Triton N-101, the inhomogeneity vanished: 95% of the monomers were involved in the P450-->P420 transition (delta V degrees = -86 ml/mol) followed by intense bleaching of the hemoprotein. Triton N 101 48-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-135 7483668-9 1995 This provides strong evidence that P450 oxidatively dealkylates the amines by a hydrogen atom transfer mechanism and not by an electron/proton transfer mechanism. Amines 68-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 8573814-5 1995 In addition, glucose metabolism, urea synthesis, P450 dependent verapamil metabolism using high performance liquid chromatography, and interleukin-6 induced "acute phase" reaction by sulfodesoxysalicylic acid-polyacrylamide gel electrophoresis detected changes of albumin synthesis were determined in primary hepatocytes and in established liver cells. Verapamil 64-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 7483668-9 1995 This provides strong evidence that P450 oxidatively dealkylates the amines by a hydrogen atom transfer mechanism and not by an electron/proton transfer mechanism. Hydrogen 80-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 7483668-0 1995 On the mechanism of amine oxidations by P450. Amines 20-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-44 7788866-0 1995 Non-specific inhibition of cytochrome P450 activities by chlorophyllin in human and rat liver microsomes. chlorophyllin 57-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 7483668-2 1995 Experimental data previously used to support an electron/proton transfer mechanism for oxidative dealkylation of amines by P450 are critically analysed with the conclusion that the mechanistic evidence is indecisive. Amines 113-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-127 7483668-8 1995 Isotope effect profiles measured for the demethylation of substituted NN-bis(dideuteriomethyl)anilines by four different forms of P450 were found to be experimentally indistinguishable from the hydrogen atom transfer profile, and distinctly different from the cation radical deprotonation profile. nn-bis(dideuteriomethyl)anilines 70-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-134 7788866-6 1995 Spectrally detectable P450 was also destroyed in microsomes and purified P450 in a reconstituted system in the presence of chlorophyllin and an NADPH-generating system. NADP 144-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 7788866-4 1995 We have investigated the in vitro effects of chlorophyllin on several P450 activities including ethoxyresorufin O-deethylation, benzyloxyresorufin O-debenzylation, coumarin 7-hydroxylation, 7-ethoxycourmarin O-deethylation, B[a]P 3-hydroxylation, and chlorzoxazone 6-hydroxylation. chlorophyllin 45-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 7788866-7 1995 These results suggest that the antigenotoxic effect of chlorophyllin might be due to inhibition of P450 enzymes involving bioactivation of carcinogens in addition to molecular complex formation between carcinogens and chlorophyllin. chlorophyllin 55-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-103 7788866-8 1995 Comparison of the apparent Ki for P450 inactivation with previously estimated constants for chlorophyllin-IQ complexation suggest that P450 inhibition should be the dominant mechanism of inhibition. chlorophyllin 92-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 7788866-8 1995 Comparison of the apparent Ki for P450 inactivation with previously estimated constants for chlorophyllin-IQ complexation suggest that P450 inhibition should be the dominant mechanism of inhibition. chlorophyllin 92-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-139 7788866-5 1995 Chlorophyllin non-specifically inhibited all of P450 activities observed. chlorophyllin 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 7788866-6 1995 Spectrally detectable P450 was also destroyed in microsomes and purified P450 in a reconstituted system in the presence of chlorophyllin and an NADPH-generating system. chlorophyllin 123-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 7788866-6 1995 Spectrally detectable P450 was also destroyed in microsomes and purified P450 in a reconstituted system in the presence of chlorophyllin and an NADPH-generating system. chlorophyllin 123-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-77 7788866-6 1995 Spectrally detectable P450 was also destroyed in microsomes and purified P450 in a reconstituted system in the presence of chlorophyllin and an NADPH-generating system. NADP 144-149 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-26 7721770-2 1995 The geminate rebinding kinetics of P450 are strongly affected by the presence of the camphor substrate. Camphor 85-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 7750560-0 1995 Substrate interactions in cytochrome P-450: correlation between carbon-13 nuclear magnetic resonance chemical shifts and C-O vibrational frequencies. Carbon 64-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-42 7734059-2 1995 During the oxygen reductase activity of P450, molecular oxygen is reduced to superoxide anion radicals (O2-.) Oxygen 11-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-44 8589076-0 1995 On the mechanism of nitric oxide formation upon oxidative cleavage of C = N(OH) bonds by NO-synthases and cytochromes P450. Nitric Oxide 20-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 7727410-0 1995 Denaturation of cytochrome P450 2B1 by guanidine hydrochloride and urea: evidence for a metastable intermediate state of the active site. Guanidine 39-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 7727410-0 1995 Denaturation of cytochrome P450 2B1 by guanidine hydrochloride and urea: evidence for a metastable intermediate state of the active site. Urea 67-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-31 7727410-1 1995 A metastable intermediate was found in the course of the denaturation of purified cytochrome P450 2B1 by increasing concentrations of guanidine hydrochloride (GuHCl). Guanidine 134-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 7727410-1 1995 A metastable intermediate was found in the course of the denaturation of purified cytochrome P450 2B1 by increasing concentrations of guanidine hydrochloride (GuHCl). Guanidine 159-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 7727410-4 1995 Before it becomes P420, the cytochrome can be completely reconverted to native P450 by dilution and incubation at 4 degrees C. Cytochrome P420 resulting from exposure to higher concentrations of GuHCl (> 3 M) failed to be reconverted to P450 by dilution. Guanidine 195-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 7727410-4 1995 Before it becomes P420, the cytochrome can be completely reconverted to native P450 by dilution and incubation at 4 degrees C. Cytochrome P420 resulting from exposure to higher concentrations of GuHCl (> 3 M) failed to be reconverted to P450 by dilution. Guanidine 195-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-244 7727410-5 1995 Denaturation of P450 2B1 by exposure to low concentrations of urea (< 2 M) is also completely reversible but no obvious intermediate is detectable. Urea 62-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 7727410-7 1995 As is the case with higher concentrations of GuHCl, cytochrome P450 denatured by exposure to 5 M or higher concentrations of urea is not reversible. Guanidine 45-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 7727410-7 1995 As is the case with higher concentrations of GuHCl, cytochrome P450 denatured by exposure to 5 M or higher concentrations of urea is not reversible. Urea 125-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-67 7890608-5 1995 The rate of CO binding to the total mixture of P450 3A4 conformers was increased in the presence of nifedipine and erythromycin, decreased by quinidine, testosterone, and warfarin, and unaffected by cimetidine and 17 alpha-ethynylestradiol. Nifedipine 100-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 7890608-5 1995 The rate of CO binding to the total mixture of P450 3A4 conformers was increased in the presence of nifedipine and erythromycin, decreased by quinidine, testosterone, and warfarin, and unaffected by cimetidine and 17 alpha-ethynylestradiol. Erythromycin 115-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 7890608-5 1995 The rate of CO binding to the total mixture of P450 3A4 conformers was increased in the presence of nifedipine and erythromycin, decreased by quinidine, testosterone, and warfarin, and unaffected by cimetidine and 17 alpha-ethynylestradiol. Quinidine 142-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 7890608-5 1995 The rate of CO binding to the total mixture of P450 3A4 conformers was increased in the presence of nifedipine and erythromycin, decreased by quinidine, testosterone, and warfarin, and unaffected by cimetidine and 17 alpha-ethynylestradiol. Testosterone 153-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 7890608-5 1995 The rate of CO binding to the total mixture of P450 3A4 conformers was increased in the presence of nifedipine and erythromycin, decreased by quinidine, testosterone, and warfarin, and unaffected by cimetidine and 17 alpha-ethynylestradiol. Warfarin 171-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 7890608-5 1995 The rate of CO binding to the total mixture of P450 3A4 conformers was increased in the presence of nifedipine and erythromycin, decreased by quinidine, testosterone, and warfarin, and unaffected by cimetidine and 17 alpha-ethynylestradiol. Ethinyl Estradiol 214-239 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-51 7832304-0 1995 Metabolism of a new local anesthetic, ropivacaine, by human hepatic cytochrome P450. Ropivacaine 38-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-83 7832304-3 1995 Because ropivacaine is an amide-type local anesthetic, it is metabolized by cytochrome P450 (P450) in the liver, and its elimination and plasma concentration can be dependent on the level of P450. Ropivacaine 8-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 7832304-3 1995 Because ropivacaine is an amide-type local anesthetic, it is metabolized by cytochrome P450 (P450) in the liver, and its elimination and plasma concentration can be dependent on the level of P450. Ropivacaine 8-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 7832304-3 1995 Because ropivacaine is an amide-type local anesthetic, it is metabolized by cytochrome P450 (P450) in the liver, and its elimination and plasma concentration can be dependent on the level of P450. Ropivacaine 8-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 7832304-3 1995 Because ropivacaine is an amide-type local anesthetic, it is metabolized by cytochrome P450 (P450) in the liver, and its elimination and plasma concentration can be dependent on the level of P450. Amides 26-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 7832304-3 1995 Because ropivacaine is an amide-type local anesthetic, it is metabolized by cytochrome P450 (P450) in the liver, and its elimination and plasma concentration can be dependent on the level of P450. Amides 26-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 7832304-3 1995 Because ropivacaine is an amide-type local anesthetic, it is metabolized by cytochrome P450 (P450) in the liver, and its elimination and plasma concentration can be dependent on the level of P450. Amides 26-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 7832304-4 1995 The purpose of this investigation was to elucidate the metabolism of ropivacaine by human hepatic P450. Ropivacaine 69-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 7832304-5 1995 METHODS: The metabolism of ropivacaine was compared using recombinant human and purified rat hepatic P450 isozymes. Ropivacaine 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 7832304-6 1995 An inhibition study using antibodies against rat P450 was performed using hepatic microsomes from human and rat to identify which P450s are involved in ropivacaine metabolism. Ropivacaine 152-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 7832304-16 1995 CONCLUSIONS: Ropivacaine was metabolized to PPX, 3"-OH Rop, and 4"-OH Rop by hepatic P450. Ropivacaine 13-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 7734059-2 1995 During the oxygen reductase activity of P450, molecular oxygen is reduced to superoxide anion radicals (O2-.) Superoxides 77-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-44 7734059-2 1995 During the oxygen reductase activity of P450, molecular oxygen is reduced to superoxide anion radicals (O2-.) Oxygen 104-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-44 7734059-3 1995 most likely by autooxidation of a P450 ferric-dioxyanion complex. ferric-dioxyanion 39-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 8589076-1 1995 Microsomal liver cytochromes P450 catalyze the oxidative cleavage of the C = NOH bond of many ketoximes, amidoximes and guanidoximes, and NO synthases catalyze the oxidation of N omega-hydroxy-L-arginine to citrulline and NO. amidoxime 105-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 7734059-6 1995 During the xenobiotic reductase activity of P450, xenobiotics are reduced by the ferrous xenobiotic complex. ferrous xenobiotic 81-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 8589076-1 1995 Microsomal liver cytochromes P450 catalyze the oxidative cleavage of the C = NOH bond of many ketoximes, amidoximes and guanidoximes, and NO synthases catalyze the oxidation of N omega-hydroxy-L-arginine to citrulline and NO. guanidoximes 120-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 7734059-10 1995 Moreover, description of the molecular mechanisms of xenobiotic and oxygen reduction reactions by P450 is limited by the lack of knowledge of the three-dimensional (3D) structure of the mammalian P450 proteins. Oxygen 68-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 7734059-10 1995 Moreover, description of the molecular mechanisms of xenobiotic and oxygen reduction reactions by P450 is limited by the lack of knowledge of the three-dimensional (3D) structure of the mammalian P450 proteins. Oxygen 68-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 196-200 8589076-1 1995 Microsomal liver cytochromes P450 catalyze the oxidative cleavage of the C = NOH bond of many ketoximes, amidoximes and guanidoximes, and NO synthases catalyze the oxidation of N omega-hydroxy-L-arginine to citrulline and NO. N(omega)-hydroxyarginine 177-203 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 7720526-6 1995 Two samples in which isoflurane potentiated CDE metabolism to the greatest rates had higher coumarin 7-hydroxylase (indicative of CYP2A6), 7-ethoxycoumarin O-deethylase (CYP2B6), and nifedipine oxidase (CYP3A4) activities than the other eight samples. Isoflurane 21-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 170-176 8589076-1 1995 Microsomal liver cytochromes P450 catalyze the oxidative cleavage of the C = NOH bond of many ketoximes, amidoximes and guanidoximes, and NO synthases catalyze the oxidation of N omega-hydroxy-L-arginine to citrulline and NO. Citrulline 207-217 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-33 8589076-3 1995 This leads to a unifying view of the mechanisms of P450- and NOS-dependent oxidative cleavage of C = NOH bonds, the relative contribution of Fe(II) O2.- being very different in NO-synthase and cytochromes P450. fe(ii) o2 141-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-64 8589076-3 1995 This leads to a unifying view of the mechanisms of P450- and NOS-dependent oxidative cleavage of C = NOH bonds, the relative contribution of Fe(II) O2.- being very different in NO-synthase and cytochromes P450. fe(ii) o2 141-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 7720526-8 1995 In microsomes from cells transfected with cDNAs coding for individual human P450s, CDE metabolism by CYP2B6 was stimulated (216%) by isoflurane, whereas isoflurane did not stimulate CDE metabolism by human CYP2A6, CYP3A4, CYP2D6, or CYP2E1. Isoflurane 133-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 19698378-3 1995 In particular, activities of the P450-IID6 isoenzyme are related to the sparteine/debrisoquine oxidation polymorphism. Sparteine 72-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-42 8820861-3 1995 The types of anthocyanins synthesised in plants are controlled by the cytochrome P450 enzymes flavonoid 3"-hydroxylase and flavonoid 3",5"-hydroxylase. Anthocyanins 13-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 8820861-3 1995 The types of anthocyanins synthesised in plants are controlled by the cytochrome P450 enzymes flavonoid 3"-hydroxylase and flavonoid 3",5"-hydroxylase. Flavonoids 94-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 7813099-4 1995 Although conjugation reactions may occasionally produce potential immunogens (for example, the generation of acylglucuronides from non-steroidal anti-inflammatory drugs such as diclofenac), bioactivation is catalysed most frequently by cytochrome P450 (P450) enzymes. acylglucuronides 109-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 253-257 7813099-4 1995 Although conjugation reactions may occasionally produce potential immunogens (for example, the generation of acylglucuronides from non-steroidal anti-inflammatory drugs such as diclofenac), bioactivation is catalysed most frequently by cytochrome P450 (P450) enzymes. Diclofenac 177-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 247-251 7813099-4 1995 Although conjugation reactions may occasionally produce potential immunogens (for example, the generation of acylglucuronides from non-steroidal anti-inflammatory drugs such as diclofenac), bioactivation is catalysed most frequently by cytochrome P450 (P450) enzymes. Diclofenac 177-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 253-257 19698378-3 1995 In particular, activities of the P450-IID6 isoenzyme are related to the sparteine/debrisoquine oxidation polymorphism. Debrisoquin 82-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-42 7696528-0 1994 The oxidation of tetrasubstituted alkenes by cytochrome P450. tetrasubstituted 17-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 7696548-6 1994 Of the 12 human P450 forms studied, CYP2B6 and CYP2E1 existing in human liver and/or lungs and CYP2F1 in human lungs were the most active in the forming of styrene glycol, followed by CYP1A2 and CYP2C8. styrene glycol 156-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 7696528-0 1994 The oxidation of tetrasubstituted alkenes by cytochrome P450. Alkenes 34-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 7696528-1 1994 Hexamethyl(Dewar benzene) (HMDB) was selected to probe the intermediacy of an alkene radical cation during cytochrome P450 (P450) catalyzed epoxidation. hexamethyl(dewar benzene) 0-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 7696528-1 1994 Hexamethyl(Dewar benzene) (HMDB) was selected to probe the intermediacy of an alkene radical cation during cytochrome P450 (P450) catalyzed epoxidation. hexamethyl(dewar benzene) 0-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 7696528-1 1994 Hexamethyl(Dewar benzene) (HMDB) was selected to probe the intermediacy of an alkene radical cation during cytochrome P450 (P450) catalyzed epoxidation. hmdb 27-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 7696528-1 1994 Hexamethyl(Dewar benzene) (HMDB) was selected to probe the intermediacy of an alkene radical cation during cytochrome P450 (P450) catalyzed epoxidation. hmdb 27-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 7696528-1 1994 Hexamethyl(Dewar benzene) (HMDB) was selected to probe the intermediacy of an alkene radical cation during cytochrome P450 (P450) catalyzed epoxidation. Alkenes 78-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-122 7696528-1 1994 Hexamethyl(Dewar benzene) (HMDB) was selected to probe the intermediacy of an alkene radical cation during cytochrome P450 (P450) catalyzed epoxidation. Alkenes 78-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 7696528-2 1994 P450 catalyzed the allylic oxidation of HMDB exclusively; no rearranged products (indicative of a radical cation intermediate) nor epoxide was detected. hmdb 40-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 7696528-3 1994 In addition, P450 exclusively catalyzed the allylic oxidation of 1,2-dimethylcyclohexene (DMCH) and 1,2,4,5-tetramethyl-1,4- cyclohexadiene. 1,2-DIMETHYLCYCLOHEXENE 65-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 7696528-3 1994 In addition, P450 exclusively catalyzed the allylic oxidation of 1,2-dimethylcyclohexene (DMCH) and 1,2,4,5-tetramethyl-1,4- cyclohexadiene. dmch 90-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 7696528-3 1994 In addition, P450 exclusively catalyzed the allylic oxidation of 1,2-dimethylcyclohexene (DMCH) and 1,2,4,5-tetramethyl-1,4- cyclohexadiene. 1,2,4,5-tetramethyl-1,4- cyclohexadiene 100-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-17 7696528-5 1994 The rates of alkene epoxidation by model systems tend to increase with increasing alkene substitution, but it is apparent that P450 has additional mechanistic options or constraints that depress the rate of epoxidation with increasing alkene substitution. Alkenes 13-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 7696528-5 1994 The rates of alkene epoxidation by model systems tend to increase with increasing alkene substitution, but it is apparent that P450 has additional mechanistic options or constraints that depress the rate of epoxidation with increasing alkene substitution. Alkenes 82-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 7696528-5 1994 The rates of alkene epoxidation by model systems tend to increase with increasing alkene substitution, but it is apparent that P450 has additional mechanistic options or constraints that depress the rate of epoxidation with increasing alkene substitution. Alkenes 82-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 7696528-6 1994 The failure of P450 to epoxidize tetrasubstituted alkenes illustrates a failure of P450 model systems to truly mimic the chemistry of the P450 iron-oxo species. tetrasubstituted 33-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 7696528-6 1994 The failure of P450 to epoxidize tetrasubstituted alkenes illustrates a failure of P450 model systems to truly mimic the chemistry of the P450 iron-oxo species. Alkenes 50-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 8093105-1 1994 The O-dealkylations of ethoxyresorufin and pentoxyresorufin are widely used activity probes for measuring the cytochrome P450 forms, CYP1A1 and CYP2B1, respectively, and their induction by xenobiotics, but there is confusion in the literature about which P450 forms are detected in human and rat liver microsomes by these and homologous alkoxyresorufins. ethoxyresorufin 23-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 8093105-10 1994 Pentoxyresorufin and benzyloxyresorufin were metabolised by several different P450 forms in non-induced rat liver microsomes but mainly by the CYP1A subfamily in 3MC-induced microsomes and by CYP2B1 in PB- and ISF-induced microsomes. pentoxyresorufin 0-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 7935340-0 1994 Bufuralol hydroxylation by cytochrome P450 2D6 and 1A2 enzymes in human liver microsomes. bufuralol 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 7935340-2 1994 In the present study we further examined the roles of several human P450 enzymes, as well as P450 2D6, in the hydroxylation of (+/-)-bufuralol, using liver microsomes from several human samples and human P450 enzymes expressed in human lymphoblastoid cell lines or Escherichia coli. bufuralol 127-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 7935340-2 1994 In the present study we further examined the roles of several human P450 enzymes, as well as P450 2D6, in the hydroxylation of (+/-)-bufuralol, using liver microsomes from several human samples and human P450 enzymes expressed in human lymphoblastoid cell lines or Escherichia coli. bufuralol 127-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 7935340-2 1994 In the present study we further examined the roles of several human P450 enzymes, as well as P450 2D6, in the hydroxylation of (+/-)-bufuralol, using liver microsomes from several human samples and human P450 enzymes expressed in human lymphoblastoid cell lines or Escherichia coli. bufuralol 127-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-97 7935340-4 1994 Quinidine and anti-rat P450 2D1 antibody almost completely inhibited bufuralol 1"-hydroxylation in human sample HL-18 at a substrate concentration of 0.4 mM, whereas these effects were not so drastic when liver microsomes from human sample HL-67 were used. bufuralol 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 7935340-6 1994 When the relative contents of P450 2D6 and P450 1A2 in 20 human samples were determined, bufuralol 1"-hydroxylation in samples containing large amounts of P450 2D6 tended to be more sensitive to quinidine, whereas the P450 1A2-rich samples were highly susceptible to alpha-naphthoflavone. bufuralol 89-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 7935340-6 1994 When the relative contents of P450 2D6 and P450 1A2 in 20 human samples were determined, bufuralol 1"-hydroxylation in samples containing large amounts of P450 2D6 tended to be more sensitive to quinidine, whereas the P450 1A2-rich samples were highly susceptible to alpha-naphthoflavone. bufuralol 89-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 7935340-6 1994 When the relative contents of P450 2D6 and P450 1A2 in 20 human samples were determined, bufuralol 1"-hydroxylation in samples containing large amounts of P450 2D6 tended to be more sensitive to quinidine, whereas the P450 1A2-rich samples were highly susceptible to alpha-naphthoflavone. bufuralol 89-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 7935340-6 1994 When the relative contents of P450 2D6 and P450 1A2 in 20 human samples were determined, bufuralol 1"-hydroxylation in samples containing large amounts of P450 2D6 tended to be more sensitive to quinidine, whereas the P450 1A2-rich samples were highly susceptible to alpha-naphthoflavone. bufuralol 89-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 7935340-7 1994 However, at low substrate concentrations bufuralol 1"-hydroxylation was shown to be catalyzed principally by P450 2D6, based on the inhibitory effects of anti-rat P450 2D1 antibody and quinidine, in both human samples HL-18 and HL-67. bufuralol 41-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 7935340-7 1994 However, at low substrate concentrations bufuralol 1"-hydroxylation was shown to be catalyzed principally by P450 2D6, based on the inhibitory effects of anti-rat P450 2D1 antibody and quinidine, in both human samples HL-18 and HL-67. bufuralol 41-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-167 7935340-7 1994 However, at low substrate concentrations bufuralol 1"-hydroxylation was shown to be catalyzed principally by P450 2D6, based on the inhibitory effects of anti-rat P450 2D1 antibody and quinidine, in both human samples HL-18 and HL-67. Quinidine 185-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-113 7935340-10 1994 The formation of the 4- and 6-hydroxylated products was suggested to be catalyzed by P450 1A2, based on the results of correlation with P450 1A2 contents in 60 human samples and inhibition by anti-P450 1A2 and alpha-naphthoflavone. alpha-naphthoflavone 210-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 7935340-10 1994 The formation of the 4- and 6-hydroxylated products was suggested to be catalyzed by P450 1A2, based on the results of correlation with P450 1A2 contents in 60 human samples and inhibition by anti-P450 1A2 and alpha-naphthoflavone. alpha-naphthoflavone 210-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 7935340-10 1994 The formation of the 4- and 6-hydroxylated products was suggested to be catalyzed by P450 1A2, based on the results of correlation with P450 1A2 contents in 60 human samples and inhibition by anti-P450 1A2 and alpha-naphthoflavone. alpha-naphthoflavone 210-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-140 7935340-11 1994 Purified recombinant P450 1A2 (expressed in E. coli) produced 1"-, 4-, and 6-hydroxybufuralol in a reconstituted system, although P450 2D6 (expressed in human lymphoblast cell lines) was found to catalyze only bufuralol 1"-hydroxylation. 1"-, 4-, and 6-hydroxybufuralol 62-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 7935340-11 1994 Purified recombinant P450 1A2 (expressed in E. coli) produced 1"-, 4-, and 6-hydroxybufuralol in a reconstituted system, although P450 2D6 (expressed in human lymphoblast cell lines) was found to catalyze only bufuralol 1"-hydroxylation. bufuralol 84-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-25 8093105-3 1994 The purified 3-methylcholanthrene (3MC)-induced rat P450 forms, CYP1A1 and CYP1A2, and a possible variant form, CYP1A1*, showed substrate selectivities for propoxyresorufin, methoxyresorufin and ethoxyresorufin, respectively. propoxyresorufin 156-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 8093105-3 1994 The purified 3-methylcholanthrene (3MC)-induced rat P450 forms, CYP1A1 and CYP1A2, and a possible variant form, CYP1A1*, showed substrate selectivities for propoxyresorufin, methoxyresorufin and ethoxyresorufin, respectively. 7-methoxyresorufin 174-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 8093105-3 1994 The purified 3-methylcholanthrene (3MC)-induced rat P450 forms, CYP1A1 and CYP1A2, and a possible variant form, CYP1A1*, showed substrate selectivities for propoxyresorufin, methoxyresorufin and ethoxyresorufin, respectively. ethoxyresorufin 175-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 8093105-8 1994 Anti-P450 antibody and furafylline inhibition of rat liver microsomal AROD confirmed that ethoxyresorufin was a selective probe for CYP1A1 in 3MC-induced and isosafrole (ISF)-induced microsomes and that pentoxy- and benzyloxyresorufins both selectively measured CYP2B1 in PB-induced and ISF-induced microsomes. ethoxyresorufin 90-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 8093105-10 1994 Pentoxyresorufin and benzyloxyresorufin were metabolised by several different P450 forms in non-induced rat liver microsomes but mainly by the CYP1A subfamily in 3MC-induced microsomes and by CYP2B1 in PB- and ISF-induced microsomes. benzyloxyresorufin 21-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 8093105-1 1994 The O-dealkylations of ethoxyresorufin and pentoxyresorufin are widely used activity probes for measuring the cytochrome P450 forms, CYP1A1 and CYP2B1, respectively, and their induction by xenobiotics, but there is confusion in the literature about which P450 forms are detected in human and rat liver microsomes by these and homologous alkoxyresorufins. ethoxyresorufin 23-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 255-259 8093105-1 1994 The O-dealkylations of ethoxyresorufin and pentoxyresorufin are widely used activity probes for measuring the cytochrome P450 forms, CYP1A1 and CYP2B1, respectively, and their induction by xenobiotics, but there is confusion in the literature about which P450 forms are detected in human and rat liver microsomes by these and homologous alkoxyresorufins. pentoxyresorufin 43-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 8093105-1 1994 The O-dealkylations of ethoxyresorufin and pentoxyresorufin are widely used activity probes for measuring the cytochrome P450 forms, CYP1A1 and CYP2B1, respectively, and their induction by xenobiotics, but there is confusion in the literature about which P450 forms are detected in human and rat liver microsomes by these and homologous alkoxyresorufins. pentoxyresorufin 43-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 255-259 8093105-1 1994 The O-dealkylations of ethoxyresorufin and pentoxyresorufin are widely used activity probes for measuring the cytochrome P450 forms, CYP1A1 and CYP2B1, respectively, and their induction by xenobiotics, but there is confusion in the literature about which P450 forms are detected in human and rat liver microsomes by these and homologous alkoxyresorufins. alkoxyresorufins 337-353 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 8093105-3 1994 The purified 3-methylcholanthrene (3MC)-induced rat P450 forms, CYP1A1 and CYP1A2, and a possible variant form, CYP1A1*, showed substrate selectivities for propoxyresorufin, methoxyresorufin and ethoxyresorufin, respectively. Methylcholanthrene 13-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 8093105-3 1994 The purified 3-methylcholanthrene (3MC)-induced rat P450 forms, CYP1A1 and CYP1A2, and a possible variant form, CYP1A1*, showed substrate selectivities for propoxyresorufin, methoxyresorufin and ethoxyresorufin, respectively. Methylcholanthrene 35-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 7913410-3 1994 Metabolite B and 6 alpha-hydroxytaxol are shown to be products of different, highly regioselective cytochrome P-450 (P450) enzymes, while metabolite A results from stepwise metabolism by each of these enzymes. alpha-hydroxytaxol 19-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-121 8055971-2 1994 The interaction between rat and human liver cytochromes P450 with a series of lysergic acid derivatives and ergopeptide alkaloids was studied by difference visible spectroscopy. Lysergic Acid 78-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 8055971-2 1994 The interaction between rat and human liver cytochromes P450 with a series of lysergic acid derivatives and ergopeptide alkaloids was studied by difference visible spectroscopy. Alkaloids 120-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-60 8055971-10 1994 Liver microsomes from rats pretreated with dexamethasone, a specific inducer of P450 3A, were found to be particularly active for the hydroxylation of bromocriptine, which occurs at the level of its tripeptide moiety. Dexamethasone 43-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 8055971-10 1994 Liver microsomes from rats pretreated with dexamethasone, a specific inducer of P450 3A, were found to be particularly active for the hydroxylation of bromocriptine, which occurs at the level of its tripeptide moiety. Bromocriptine 151-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 8055971-10 1994 Liver microsomes from rats pretreated with dexamethasone, a specific inducer of P450 3A, were found to be particularly active for the hydroxylation of bromocriptine, which occurs at the level of its tripeptide moiety. tripeptide K-26 199-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-84 8055971-11 1994 Human liver microsomes as well as P450 NF25 and P450 hPCN1 also exhibited a high activity for bromocriptine hydroxylation at this level. Bromocriptine 94-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-38 8055971-11 1994 Human liver microsomes as well as P450 NF25 and P450 hPCN1 also exhibited a high activity for bromocriptine hydroxylation at this level. Bromocriptine 94-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 8055971-13 1994 The tripeptide moiety of ergopeptides is essential for their recognition by P450s 3A and binds at a site close to P450 heme, producing type-I difference spectra. tripeptide K-26 4-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 7819404-1 1994 Administration of a perfluorodecalin (PFD) emulsion, the liver cytochrome P-450 II B1/B2 inducer, to experimental animals is followed by a two-fold increase of the NADPH oxidation rate in liver microsomes. perfluorodecalin 20-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-88 7819404-1 1994 Administration of a perfluorodecalin (PFD) emulsion, the liver cytochrome P-450 II B1/B2 inducer, to experimental animals is followed by a two-fold increase of the NADPH oxidation rate in liver microsomes. perfluorodecalin 38-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-88 7819404-1 1994 Administration of a perfluorodecalin (PFD) emulsion, the liver cytochrome P-450 II B1/B2 inducer, to experimental animals is followed by a two-fold increase of the NADPH oxidation rate in liver microsomes. NADP 164-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-88 8021301-4 1994 After an incubation of the cells with radiolabeled proline or methionine, two major proteins were identified, p450-480 and p290 (so named because of their molecular masses). Proline 51-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-114 8021301-4 1994 After an incubation of the cells with radiolabeled proline or methionine, two major proteins were identified, p450-480 and p290 (so named because of their molecular masses). Methionine 62-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-114 8021301-6 1994 P450-480 resolved to M(r) 190,000 under reducing sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) conditions. Sodium Dodecyl Sulfate 49-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 8021301-6 1994 P450-480 resolved to M(r) 190,000 under reducing sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) conditions. polyacrylamide gels 72-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 8021301-6 1994 P450-480 resolved to M(r) 190,000 under reducing sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) conditions. Sodium Dodecyl Sulfate 108-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-4 8296319-7 1994 The similarities of P450- and Hb-mediated aniline hydroxylation using stable isotopes preclude their use as in vivo probes. aniline 42-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-32 8203907-0 1994 Evaluation of triacetyloleandomycin, alpha-naphthoflavone and diethyldithiocarbamate as selective chemical probes for inhibition of human cytochromes P450. Troleandomycin 14-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 8203907-0 1994 Evaluation of triacetyloleandomycin, alpha-naphthoflavone and diethyldithiocarbamate as selective chemical probes for inhibition of human cytochromes P450. alpha-naphthoflavone 37-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 8203907-0 1994 Evaluation of triacetyloleandomycin, alpha-naphthoflavone and diethyldithiocarbamate as selective chemical probes for inhibition of human cytochromes P450. Ditiocarb 62-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-154 8203907-7 1994 Decreasing the concentration of DDC to 10 microM, however, led to inhibition of CYP2A6 (65% inhibition) and CYP2B6 (50% inhibition), but none of the other P450s examined, including CYP2E1. Ditiocarb 32-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 108-114 7517356-1 1994 Membrane-bound sites of cytochrome P-450 2B4 (LM2) were determined by means of two different methods, photoactivated binding of membrane phospholipids to the protein and epitope mapping by antibodies. Phospholipids 137-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-40 8079499-0 1994 Interaction of a series of nitriles with the alcohol-inducible isoform of P450: computer analysis of structure-activity relationships. Nitriles 27-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-78 8079499-0 1994 Interaction of a series of nitriles with the alcohol-inducible isoform of P450: computer analysis of structure-activity relationships. Alcohols 45-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-78 8117105-7 1994 Human P450 1A2 was solubilized using high concentrations of sodium cholate and Triton N-101 and could be purified to near homogeneity in high yield in two steps. Sodium Cholate 60-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-10 8117105-7 1994 Human P450 1A2 was solubilized using high concentrations of sodium cholate and Triton N-101 and could be purified to near homogeneity in high yield in two steps. Triton N 101 79-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-10 8117105-9 1994 The purified human P450 1A2 was found to be almost completely in the high spin iron configuration, in contrast to P450 1A2 enzymes isolated from rats and rabbits. Iron 79-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-23 7945891-7 1994 Aromatase and cholesterol side-chain cleaving P450 mRNAs, proteins, and activities are measurable in human placentas and do not seem to be affected by maternal cigarette smoking. Cholesterol 14-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-50 8286335-1 1994 Oxidation of tienilic acid (TA) by microsomes of yeast expressing two closely related human liver cytochrome P-450s (P450), P450 2C9 and 2C10, led to catalysis-dependent loss of activity of these P450s. Ticrynafen 13-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-121 8974327-3 1994 The enzyme constitutes the only P450 form that is strongly induced by ethanol. Ethanol 70-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 8283039-2 1994 Of these, Fc gamma RIIb1* represents a receptor variant that is identical to IIb1 except for a single amino acid difference in the cytoplasmic tail (amino acid position 11) where a tyrosine (IIb1) is replaced by an aspartic acid (IIb1*). Tyrosine 181-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 8283039-2 1994 Of these, Fc gamma RIIb1* represents a receptor variant that is identical to IIb1 except for a single amino acid difference in the cytoplasmic tail (amino acid position 11) where a tyrosine (IIb1) is replaced by an aspartic acid (IIb1*). Aspartic Acid 215-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 8286335-1 1994 Oxidation of tienilic acid (TA) by microsomes of yeast expressing two closely related human liver cytochrome P-450s (P450), P450 2C9 and 2C10, led to catalysis-dependent loss of activity of these P450s. Ticrynafen 13-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 8286335-1 1994 Oxidation of tienilic acid (TA) by microsomes of yeast expressing two closely related human liver cytochrome P-450s (P450), P450 2C9 and 2C10, led to catalysis-dependent loss of activity of these P450s. Ticrynafen 28-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-121 8286335-1 1994 Oxidation of tienilic acid (TA) by microsomes of yeast expressing two closely related human liver cytochrome P-450s (P450), P450 2C9 and 2C10, led to catalysis-dependent loss of activity of these P450s. Ticrynafen 28-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 8286335-3 1994 The loss of P450 activity during TA oxidation was concomitant with product (5-hydroxytienilic acid, 5-OHTA) formation, showed pseudo-first-order and saturation kinetics, and was inhibited by an alternative substrate, tolbutamide. 5-hydroxythienilic acid 76-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-16 8286335-3 1994 The loss of P450 activity during TA oxidation was concomitant with product (5-hydroxytienilic acid, 5-OHTA) formation, showed pseudo-first-order and saturation kinetics, and was inhibited by an alternative substrate, tolbutamide. 5-ohta 100-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-16 8286335-3 1994 The loss of P450 activity during TA oxidation was concomitant with product (5-hydroxytienilic acid, 5-OHTA) formation, showed pseudo-first-order and saturation kinetics, and was inhibited by an alternative substrate, tolbutamide. Tolbutamide 217-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-16 8286335-8 1994 Moreover, a specific covalent binding of 0.9 mol of TA metabolite per mole of P450 2C10 was found to occur before the complete loss of enzyme activity (in incubations performed in the presence of glutathione). Glutathione 196-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 8286335-10 1994 It involves the intermediate formation of an electrophilic thiophene sulfoxide, which may react at position 5 of its thiophene ring either with H2O to give 5-OHTA or with a nucleophilic group of an amino acid residue of the P450 active site, which results in its covalent binding to P450 protein. thiophene sulfoxide 59-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 224-228 8286335-10 1994 It involves the intermediate formation of an electrophilic thiophene sulfoxide, which may react at position 5 of its thiophene ring either with H2O to give 5-OHTA or with a nucleophilic group of an amino acid residue of the P450 active site, which results in its covalent binding to P450 protein. thiophene sulfoxide 59-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 283-287 8286335-10 1994 It involves the intermediate formation of an electrophilic thiophene sulfoxide, which may react at position 5 of its thiophene ring either with H2O to give 5-OHTA or with a nucleophilic group of an amino acid residue of the P450 active site, which results in its covalent binding to P450 protein. Thiophenes 59-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 224-228 8286335-10 1994 It involves the intermediate formation of an electrophilic thiophene sulfoxide, which may react at position 5 of its thiophene ring either with H2O to give 5-OHTA or with a nucleophilic group of an amino acid residue of the P450 active site, which results in its covalent binding to P450 protein. Thiophenes 59-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 283-287 8286335-10 1994 It involves the intermediate formation of an electrophilic thiophene sulfoxide, which may react at position 5 of its thiophene ring either with H2O to give 5-OHTA or with a nucleophilic group of an amino acid residue of the P450 active site, which results in its covalent binding to P450 protein. Water 144-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 224-228 8286335-10 1994 It involves the intermediate formation of an electrophilic thiophene sulfoxide, which may react at position 5 of its thiophene ring either with H2O to give 5-OHTA or with a nucleophilic group of an amino acid residue of the P450 active site, which results in its covalent binding to P450 protein. Water 144-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 283-287 8286335-10 1994 It involves the intermediate formation of an electrophilic thiophene sulfoxide, which may react at position 5 of its thiophene ring either with H2O to give 5-OHTA or with a nucleophilic group of an amino acid residue of the P450 active site, which results in its covalent binding to P450 protein. 5-ohta 156-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 224-228 8286335-10 1994 It involves the intermediate formation of an electrophilic thiophene sulfoxide, which may react at position 5 of its thiophene ring either with H2O to give 5-OHTA or with a nucleophilic group of an amino acid residue of the P450 active site, which results in its covalent binding to P450 protein. 5-ohta 156-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 283-287 8274159-0 1993 Cytochrome P450 dependent N-hydroxylation of a guanidine (debrisoquine), microsomal catalysed reduction and further oxidation of the N-hydroxy-guanidine metabolite to the urea derivative. Guanidine 47-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 8022846-0 1994 Oxygenation of polyunsaturated fatty acids by cytochrome P450 monooxygenases. Fatty Acids, Unsaturated 15-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8022846-1 1994 Polyunsaturated fatty acids can be oxygenated by P450 in different ways--by epoxidation, by hydroxylation of the omega-side chain, by allylic and bis-allylic hydroxylation and by hydroxylation with double bond migration. Fatty Acids, Unsaturated 0-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 8022846-5 1994 Many observations indicate that P450 oxygenates arachidonic acid in vivo in man and in experimental animals. Arachidonic Acid 48-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-36 8022846-8 1994 It was more unexpected that biological activities have been associated with many of the P450 metabolites of arachidonic acid, at least in pharmacological doses. Arachidonic Acid 108-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 8022846-10 1994 In their critical review on epoxygenase metabolism of arachidonic acid in 1989, Fitzpatrick and Murphy pointed out some major differences between the PGH synthase, the lipoxygenase and the P450 pathways of arachidonic acid metabolism. Arachidonic Acid 54-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-193 8022846-10 1994 In their critical review on epoxygenase metabolism of arachidonic acid in 1989, Fitzpatrick and Murphy pointed out some major differences between the PGH synthase, the lipoxygenase and the P450 pathways of arachidonic acid metabolism. Arachidonic Acid 206-222 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-193 8274159-0 1993 Cytochrome P450 dependent N-hydroxylation of a guanidine (debrisoquine), microsomal catalysed reduction and further oxidation of the N-hydroxy-guanidine metabolite to the urea derivative. Nitrogen 26-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 8274159-0 1993 Cytochrome P450 dependent N-hydroxylation of a guanidine (debrisoquine), microsomal catalysed reduction and further oxidation of the N-hydroxy-guanidine metabolite to the urea derivative. Debrisoquin 58-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 8274159-0 1993 Cytochrome P450 dependent N-hydroxylation of a guanidine (debrisoquine), microsomal catalysed reduction and further oxidation of the N-hydroxy-guanidine metabolite to the urea derivative. hydroxyguanidine 133-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 8274159-0 1993 Cytochrome P450 dependent N-hydroxylation of a guanidine (debrisoquine), microsomal catalysed reduction and further oxidation of the N-hydroxy-guanidine metabolite to the urea derivative. Urea 171-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 8274159-6 1993 The transformation of N-hydroxydebrisoquine to the corresponding urea derivative was also detected in in vitro experiments with microsomal fractions and enriched P450 fractions as well as with flavin-containing monooxygenase (FMO). N-hydroxydebrisoquine 22-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-166 8274159-6 1993 The transformation of N-hydroxydebrisoquine to the corresponding urea derivative was also detected in in vitro experiments with microsomal fractions and enriched P450 fractions as well as with flavin-containing monooxygenase (FMO). Urea 65-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-166 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Nitrogen 12-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Nitrogen 12-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 176-180 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Guanidine 35-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Guanidine 35-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 176-180 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Hydrogen Peroxide 195-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Hydrogen Peroxide 195-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 176-180 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Oxygen 197-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Oxygen 197-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 176-180 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. hydroxyguanidine 120-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. hydroxyguanidine 120-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 176-180 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Urea 294-298 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 8274159-8 1993 Whereas the N-hydroxylation of the guanidine involves the usual monooxygenase activity of cytochrome P450 the resultant N-hydroxyguanidine decouples monooxygenases (cytochrome P450, FMO) and the H2O2 and, above all, O2- thus formed transform the N-hydroxyguanidine further to the corresponding urea derivative. Urea 294-298 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 176-180 8269634-4 1993 Microsomal metabolism of AFB1 to AFB1 8,9-epoxide (as measured by AFB1 tris-diol formation) and aflatoxin Q1 (AFQ1), the major metabolite produced, was significantly correlated with CYP3A3/4 expression (P < 0.001) and, to a lesser extent, with CYP2B6 expression (P < 0.01). aflatoxin B1-2,3-oxide 33-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 247-253 8109933-0 1993 Ro 09-1470 is a selective inhibitor of P-450 lanosterol C-14 demethylase of fungi. restricticin 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-44 8109933-1 1993 Ro 09-1470 is a new antifungal agent that belongs to a series of compounds characterized by a tetrahydropyran skeleton with glycine and alkenyl side chains and that inhibits P-450 lanosterol C-14 demethylase (P-450(14DM)) of fungi (Y. Aoki, T. Yamazaki, M. Kondoh, Y. Sudoh, N. Nakayama, Y. Sekine, H. Shimada, and M. Arisawa, J. Antibiot. restricticin 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 174-179 8242617-13 1993 Overall, the present study establishes that liver microsomal CYP2B and CYP3A preferentially catalyze cyclophosphamide and ifosphamide 4-hydroxylation, respectively, suggesting that liver P-450-inducing agents targeted at these enzymes might be used in cancer patients to enhance drug activation and therapeutic efficacy. Cyclophosphamide 101-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-66 8242617-13 1993 Overall, the present study establishes that liver microsomal CYP2B and CYP3A preferentially catalyze cyclophosphamide and ifosphamide 4-hydroxylation, respectively, suggesting that liver P-450-inducing agents targeted at these enzymes might be used in cancer patients to enhance drug activation and therapeutic efficacy. Ifosfamide 122-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-66 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 2 subfamily B member 6 Homo sapiens 44-50 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 2 subfamily B member 6 Homo sapiens 44-50 8242617-10 1993 Orphenadrine (a CYP2B6 inhibitor) and anti-CYP2B IgG inhibited microsomal cyclophosphamide hydroxylation to a greater extent than ifosphamide hydroxylation, consistent with the 8-fold higher activity of complementary DNA-expressed CYP2B6 with cyclophosphamide. Orphenadrine 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 8242617-10 1993 Orphenadrine (a CYP2B6 inhibitor) and anti-CYP2B IgG inhibited microsomal cyclophosphamide hydroxylation to a greater extent than ifosphamide hydroxylation, consistent with the 8-fold higher activity of complementary DNA-expressed CYP2B6 with cyclophosphamide. Orphenadrine 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-21 8242617-10 1993 Orphenadrine (a CYP2B6 inhibitor) and anti-CYP2B IgG inhibited microsomal cyclophosphamide hydroxylation to a greater extent than ifosphamide hydroxylation, consistent with the 8-fold higher activity of complementary DNA-expressed CYP2B6 with cyclophosphamide. Orphenadrine 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 231-237 8242617-10 1993 Orphenadrine (a CYP2B6 inhibitor) and anti-CYP2B IgG inhibited microsomal cyclophosphamide hydroxylation to a greater extent than ifosphamide hydroxylation, consistent with the 8-fold higher activity of complementary DNA-expressed CYP2B6 with cyclophosphamide. Cyclophosphamide 74-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 8269634-4 1993 Microsomal metabolism of AFB1 to AFB1 8,9-epoxide (as measured by AFB1 tris-diol formation) and aflatoxin Q1 (AFQ1), the major metabolite produced, was significantly correlated with CYP3A3/4 expression (P < 0.001) and, to a lesser extent, with CYP2B6 expression (P < 0.01). Aflatoxins 96-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 247-253 8323289-0 1993 Role of lysine and arginine residues of cytochrome P450 in the interaction between cytochrome P4502B1 and NADPH-cytochrome P450 reductase. Lysine 8-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 8091338-11 1993 Some P450 (3A, 1A, 2E, ...) are inducible by compounds such as phenobarbital, rifampicin, aromatic hydrocarbon, ethanol, or omeprazole. Phenobarbital 63-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 8091338-11 1993 Some P450 (3A, 1A, 2E, ...) are inducible by compounds such as phenobarbital, rifampicin, aromatic hydrocarbon, ethanol, or omeprazole. Rifampin 78-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 8091338-11 1993 Some P450 (3A, 1A, 2E, ...) are inducible by compounds such as phenobarbital, rifampicin, aromatic hydrocarbon, ethanol, or omeprazole. Hydrocarbons, Aromatic 90-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 8091338-11 1993 Some P450 (3A, 1A, 2E, ...) are inducible by compounds such as phenobarbital, rifampicin, aromatic hydrocarbon, ethanol, or omeprazole. Ethanol 112-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 8091338-11 1993 Some P450 (3A, 1A, 2E, ...) are inducible by compounds such as phenobarbital, rifampicin, aromatic hydrocarbon, ethanol, or omeprazole. Omeprazole 124-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-9 8220911-0 1993 Human liver microsomal metabolism of the enantiomers of warfarin and acenocoumarol: P450 isozyme diversity determines the differences in their pharmacokinetics. Warfarin 56-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 8220911-0 1993 Human liver microsomal metabolism of the enantiomers of warfarin and acenocoumarol: P450 isozyme diversity determines the differences in their pharmacokinetics. Acenocoumarol 69-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-88 8220911-18 1993 The results suggest that the coumarin ring hydroxylations of both compounds are catalysed by different combinations of P450 isozymes. coumarin 29-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-123 8220911-19 1993 The 7-hydroxylation of R/S acenocoumarol and the 6-hydroxylation of S-acenocoumarol are at least partly conducted by (a) P450 isozyme(s) of the 2C subfamily different from P450 2C9 (the main S-warfarin 7- and 6-hydroxylase). Acenocoumarol 27-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 8220911-19 1993 The 7-hydroxylation of R/S acenocoumarol and the 6-hydroxylation of S-acenocoumarol are at least partly conducted by (a) P450 isozyme(s) of the 2C subfamily different from P450 2C9 (the main S-warfarin 7- and 6-hydroxylase). Acenocoumarol 27-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 8220911-19 1993 The 7-hydroxylation of R/S acenocoumarol and the 6-hydroxylation of S-acenocoumarol are at least partly conducted by (a) P450 isozyme(s) of the 2C subfamily different from P450 2C9 (the main S-warfarin 7- and 6-hydroxylase). Acenocoumarol 68-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 8220911-19 1993 The 7-hydroxylation of R/S acenocoumarol and the 6-hydroxylation of S-acenocoumarol are at least partly conducted by (a) P450 isozyme(s) of the 2C subfamily different from P450 2C9 (the main S-warfarin 7- and 6-hydroxylase). Acenocoumarol 68-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 172-176 8232610-0 1993 Identification of P450 enzymes involved in metabolism of verapamil in humans. Verapamil 57-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-22 8232610-6 1993 The maximum rate of formation of D-617 and norverapamil was determined in the microsomal fraction of 21 human livers which had been previously characterized for the individual expression of various P450 enzymes (CYP1A2, CYP2C, CYP2D6, CYP2E1 and CYP3A3/4) by means of Western blotting. 2-(3,4-dimethoxyphenyl)-5-amino-2-isopropylvaleronitrile 33-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-202 8232610-6 1993 The maximum rate of formation of D-617 and norverapamil was determined in the microsomal fraction of 21 human livers which had been previously characterized for the individual expression of various P450 enzymes (CYP1A2, CYP2C, CYP2D6, CYP2E1 and CYP3A3/4) by means of Western blotting. norverapamil 43-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-202 8349787-1 1993 Placenta and endometrium carry out steroidogenic biotransformation reactions such as 6-beta-hydroxylation of cortisol, a reaction characteristic of the dominant family of cytochromes P450 in human liver, CYP3A. 6-beta 85-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 8349787-1 1993 Placenta and endometrium carry out steroidogenic biotransformation reactions such as 6-beta-hydroxylation of cortisol, a reaction characteristic of the dominant family of cytochromes P450 in human liver, CYP3A. Hydrocortisone 109-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 183-187 8344304-0 1993 Kinetics of cytochromes P-450 IA1 and IIB1 in reconstituted systems with dilauroyl- and distearoyl-glycerophosphocholine. dilauroyl- and distearoyl-glycerophosphocholine 73-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-29 8344304-0 1993 Kinetics of cytochromes P-450 IA1 and IIB1 in reconstituted systems with dilauroyl- and distearoyl-glycerophosphocholine. dilauroyl- and distearoyl-glycerophosphocholine 73-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. Phosphatidylcholines 69-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-245 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. Phosphatidylcholines 69-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 254-258 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. dilauroyl 94-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-245 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. dilauroyl 94-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 254-258 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. distearoyl 107-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-245 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. distearoyl 107-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 254-258 8230303-1 1993 Cytochromes P-450 (P-450s) are a large group of heme-containing proteins that carry out oxidation of numerous chemicals. Heme 48-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-17 8230303-1 1993 Cytochromes P-450 (P-450s) are a large group of heme-containing proteins that carry out oxidation of numerous chemicals. Heme 48-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 8230303-2 1993 In mammals, a limited number of P-450s are involved in metabolic pathways of steroid synthesis, while most of these enzymes are involved in metabolism of foreign compounds. Steroids 77-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 8357281-7 1993 Fluconazole, a more selective agent for fungal P-450, seems to be of less concern regarding the potential for drug interactions than ketoconazole. Fluconazole 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-52 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. ethoxyresorufin 47-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. 7-methoxyresorufin 67-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. 7-methoxyresorufin 67-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 217-221 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. 7-methoxyresorufin 67-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 217-221 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. benz(a)anthracene 164-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. benz(a)anthracene 164-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 217-221 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. benz(a)anthracene 164-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 217-221 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. 7-methoxyresorufin 258-274 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. benzyloxyresorufin 279-297 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. pentoxyresorufin 375-391 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 8395842-3 1993 Antibody inhibition experiments indicated that ethoxyresorufin and methoxyresorufin O-dealkylations were catalysed mainly by the P450 1A subfamily in untreated and BA-induced HepG2 cells, that additional unidentified P450 forms were considerably involved in methoxyresorufin and benzyloxyresorufin O-dealkylations and that the P450 2B subfamily was partially responsible for pentoxyresorufin O-dealkylation in PB-induced cells. Phenobarbital 410-412 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 129-133 8344304-5 1993 Furthermore, the results of the present study demonstrate that the increase in the rate of O-dealkylation of ethoxycoumarin, reported in the literature for reconstituted systems in the presence of Lau2GroPCho, results from an effect of Lau2GroPCho on both the Kms and the V. In a number of additional experiments possible mechanisms underlying the observed differential effect of Lau2GroPCho and Ste2GroPCho on the Kms and V of P-450 IA1 and IIB1 were investigated. 3-ethoxychromen-2-one 109-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 428-433 8344304-5 1993 Furthermore, the results of the present study demonstrate that the increase in the rate of O-dealkylation of ethoxycoumarin, reported in the literature for reconstituted systems in the presence of Lau2GroPCho, results from an effect of Lau2GroPCho on both the Kms and the V. In a number of additional experiments possible mechanisms underlying the observed differential effect of Lau2GroPCho and Ste2GroPCho on the Kms and V of P-450 IA1 and IIB1 were investigated. 3-ethoxychromen-2-one 109-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 442-446 8344304-6 1993 This was done by studying the effect of the two acyl2GroPCho species on the kinetic parameters of some of the different steps of the P-450 cycle, namely substrate binding, oxygen binding and the rate of electron transfer. Oxygen 172-178 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-138 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. alkoxyresorufins 165-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-245 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. alkoxyresorufins 165-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 254-258 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. 3-ethoxychromen-2-one 186-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-245 8344304-1 1993 In the present study the effect of changing the fatty acyl moiety of phosphatidylcholine from dilauroyl to distearoyl on the kinetic parameters of O-dealkylation of alkoxyresorufins and ethoxycoumarin dependent on reconstituted cytochromes P-450 IA1 and IIB1 has been investigated. 3-ethoxychromen-2-one 186-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 254-258 8344304-4 1993 From these last two observations it was concluded that the mechanism by which phospholipids influence P-450-IIB1-dependent O-dealkylation of ethoxycoumarin is different from that by which they influence P-450-IIB1-dependent O-dealkylation of this substrate. Phospholipids 78-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-112 8344304-4 1993 From these last two observations it was concluded that the mechanism by which phospholipids influence P-450-IIB1-dependent O-dealkylation of ethoxycoumarin is different from that by which they influence P-450-IIB1-dependent O-dealkylation of this substrate. 3-ethoxychromen-2-one 141-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-112 8323289-0 1993 Role of lysine and arginine residues of cytochrome P450 in the interaction between cytochrome P4502B1 and NADPH-cytochrome P450 reductase. Arginine 19-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-55 8323289-1 1993 Chemical modification of cytochrome P450 was used to study the involvement of lysine and arginine residues in the interaction between cytochrome P450 and NADPH-cytochrome P450 reductase. Lysine 78-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 8323289-1 1993 Chemical modification of cytochrome P450 was used to study the involvement of lysine and arginine residues in the interaction between cytochrome P450 and NADPH-cytochrome P450 reductase. Lysine 78-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-149 8323289-1 1993 Chemical modification of cytochrome P450 was used to study the involvement of lysine and arginine residues in the interaction between cytochrome P450 and NADPH-cytochrome P450 reductase. Arginine 89-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 8323289-1 1993 Chemical modification of cytochrome P450 was used to study the involvement of lysine and arginine residues in the interaction between cytochrome P450 and NADPH-cytochrome P450 reductase. Arginine 89-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-149 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Lysine 34-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Lysine 34-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Lysine 34-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Lysine 34-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. acetic anhydride 57-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. acetic anhydride 57-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. acetic anhydride 57-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. acetic anhydride 57-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8330339-0 1993 Roles of different forms of cytochrome P450 in the activation of the promutagen 6-aminochrysene to genotoxic metabolites in human liver microsomes. 6-chrysenamine 80-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 8330339-1 1993 We reported previously that the potent mutagen 6-aminochrysene is catalyzed principally by rat liver microsomal P4501A and P4502B enzymes to reactive metabolites that induce umu gene expression in O-acetyltransferase-over-expressing strain Salmonella typhimurium NM2009; the proposal was made that there are different mechanisms in the formation of reactive N-hydroxylated and diolepoxide metabolites by P450 enzymes (Yamazaki, H. and Shimada, T., Biochem. 6-chrysenamine 47-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Benzphetamine 123-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 8330339-1 1993 We reported previously that the potent mutagen 6-aminochrysene is catalyzed principally by rat liver microsomal P4501A and P4502B enzymes to reactive metabolites that induce umu gene expression in O-acetyltransferase-over-expressing strain Salmonella typhimurium NM2009; the proposal was made that there are different mechanisms in the formation of reactive N-hydroxylated and diolepoxide metabolites by P450 enzymes (Yamazaki, H. and Shimada, T., Biochem. Nitrogen 263-264 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Benzphetamine 123-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Benzphetamine 123-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8330339-1 1993 We reported previously that the potent mutagen 6-aminochrysene is catalyzed principally by rat liver microsomal P4501A and P4502B enzymes to reactive metabolites that induce umu gene expression in O-acetyltransferase-over-expressing strain Salmonella typhimurium NM2009; the proposal was made that there are different mechanisms in the formation of reactive N-hydroxylated and diolepoxide metabolites by P450 enzymes (Yamazaki, H. and Shimada, T., Biochem. diolepoxide 377-388 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-116 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Benzphetamine 123-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8330339-3 1993 Here we further examined the roles of human liver P450 enzymes and the mechanism of activation of 6-aminochrysene by rat and human P450 enzymes in the Salmonella tester strains. 6-chrysenamine 98-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-135 8330339-6 1993 Among purified P450 enzymes examined, P4501A2 as well as P4503A4 were highly active in transforming 6-amino-chrysene to reactive metabolites, suggesting the involvement of different human P450 enzymes in the reaction. 6-chrysenamine 100-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-19 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. NADP 163-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 8330339-6 1993 Among purified P450 enzymes examined, P4501A2 as well as P4503A4 were highly active in transforming 6-amino-chrysene to reactive metabolites, suggesting the involvement of different human P450 enzymes in the reaction. 6-chrysenamine 100-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-42 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. NADP 163-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. NADP 163-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. NADP 163-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 193-197 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Lysine 256-262 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. cumene hydroperoxide 291-311 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 8323289-2 1993 Acetylation of 2.2 and 8.5 mol of lysine/mole of P450 by acetic anhydride led to 38.7 and 95% reductions, respectively, in benzphetamine demethylation activity by NADPH-dependent reconstituted P450/reductase complex, while modification of up to 8.5 mol of lysine/mol of P450 did not inhibit cumene hydroperoxide-supported P450-dependent benzphetamine demethylation. Benzphetamine 337-350 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 8323289-9 1993 These results support the hypothesis of a predominant role of lysine residues of P450 in the electrostatic interaction with NADPH-cytochrome P450 reductase. Lysine 62-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-85 8503477-12 1993 Fluoxetine, norfluoxetine (the major metabolite of fluoxetine), and paroxetine are potent inhibitors of the hepatic isoenzyme P450 IID6, whereas sertraline has much weaker inhibitory effects on its activity. Fluoxetine 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 8503477-12 1993 Fluoxetine, norfluoxetine (the major metabolite of fluoxetine), and paroxetine are potent inhibitors of the hepatic isoenzyme P450 IID6, whereas sertraline has much weaker inhibitory effects on its activity. norfluoxetine 12-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 8503477-12 1993 Fluoxetine, norfluoxetine (the major metabolite of fluoxetine), and paroxetine are potent inhibitors of the hepatic isoenzyme P450 IID6, whereas sertraline has much weaker inhibitory effects on its activity. Fluoxetine 15-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 8503477-12 1993 Fluoxetine, norfluoxetine (the major metabolite of fluoxetine), and paroxetine are potent inhibitors of the hepatic isoenzyme P450 IID6, whereas sertraline has much weaker inhibitory effects on its activity. Paroxetine 68-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 8503477-12 1993 Fluoxetine, norfluoxetine (the major metabolite of fluoxetine), and paroxetine are potent inhibitors of the hepatic isoenzyme P450 IID6, whereas sertraline has much weaker inhibitory effects on its activity. Sertraline 145-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 8503477-13 1993 Inhibition of P450 isoenzymes can cause potentially dangerous increases in the plasma levels of a large number of drugs, including TCAs, neuroleptics, and mood stabilizers, such as carbamazepine. Carbamazepine 181-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 8460938-7 1993 The monooxygenase activities of the purified human brain P450s were demonstrated with various substrates (aminopyrine, morphine, aniline, 7-ethoxycoumarin, and nifedipine) as examined in reconstituted systems consisting of purified human brain P450, purified rat brain NADPH cytochrome P450 reductase, deoxycholate, phospholipid, and NADPH. Aminopyrine 106-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8516280-8 1993 The incorporation of the integral membrane protein cytochrome P-450 into PC and PG liposomes caused a decrease in their stability towards PLA2, the decrease depending on the lipid/protein molar ratio. Phosphatidylglycerols 80-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-67 8460938-7 1993 The monooxygenase activities of the purified human brain P450s were demonstrated with various substrates (aminopyrine, morphine, aniline, 7-ethoxycoumarin, and nifedipine) as examined in reconstituted systems consisting of purified human brain P450, purified rat brain NADPH cytochrome P450 reductase, deoxycholate, phospholipid, and NADPH. Morphine 119-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8460938-7 1993 The monooxygenase activities of the purified human brain P450s were demonstrated with various substrates (aminopyrine, morphine, aniline, 7-ethoxycoumarin, and nifedipine) as examined in reconstituted systems consisting of purified human brain P450, purified rat brain NADPH cytochrome P450 reductase, deoxycholate, phospholipid, and NADPH. aniline 129-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8460938-7 1993 The monooxygenase activities of the purified human brain P450s were demonstrated with various substrates (aminopyrine, morphine, aniline, 7-ethoxycoumarin, and nifedipine) as examined in reconstituted systems consisting of purified human brain P450, purified rat brain NADPH cytochrome P450 reductase, deoxycholate, phospholipid, and NADPH. 7-ethoxycoumarin 138-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8460938-7 1993 The monooxygenase activities of the purified human brain P450s were demonstrated with various substrates (aminopyrine, morphine, aniline, 7-ethoxycoumarin, and nifedipine) as examined in reconstituted systems consisting of purified human brain P450, purified rat brain NADPH cytochrome P450 reductase, deoxycholate, phospholipid, and NADPH. Nifedipine 160-170 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8460938-7 1993 The monooxygenase activities of the purified human brain P450s were demonstrated with various substrates (aminopyrine, morphine, aniline, 7-ethoxycoumarin, and nifedipine) as examined in reconstituted systems consisting of purified human brain P450, purified rat brain NADPH cytochrome P450 reductase, deoxycholate, phospholipid, and NADPH. Deoxycholic Acid 302-314 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8460938-7 1993 The monooxygenase activities of the purified human brain P450s were demonstrated with various substrates (aminopyrine, morphine, aniline, 7-ethoxycoumarin, and nifedipine) as examined in reconstituted systems consisting of purified human brain P450, purified rat brain NADPH cytochrome P450 reductase, deoxycholate, phospholipid, and NADPH. Phospholipids 316-328 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8460938-7 1993 The monooxygenase activities of the purified human brain P450s were demonstrated with various substrates (aminopyrine, morphine, aniline, 7-ethoxycoumarin, and nifedipine) as examined in reconstituted systems consisting of purified human brain P450, purified rat brain NADPH cytochrome P450 reductase, deoxycholate, phospholipid, and NADPH. NADP 269-274 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-61 8380689-0 1993 HepG2 cells: an in vitro model for P450-dependent metabolism of acetaminophen. Acetaminophen 64-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-39 8423765-0 1993 Identification and characterization of the cytochrome P450 enzymes involved in N-dealkylation of propafenone: molecular base for interaction potential and variable disposition of active metabolites. Nitrogen 79-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 8380334-2 1993 The resultant proximal cysteine and tyrosine mutant Mbs (H93C and H93Y Mbs, respectively) exhibit the altered axial ligation analogous to P-450, chloroperoxidase, and catalase. Cysteine 23-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-175 8380334-2 1993 The resultant proximal cysteine and tyrosine mutant Mbs (H93C and H93Y Mbs, respectively) exhibit the altered axial ligation analogous to P-450, chloroperoxidase, and catalase. Tyrosine 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-175 8070855-4 1993 Of the 10 human P450 forms studied, CYP2B6 was the most effective in forming of styrene glycol, followed by CYP1A2, CYP2E1 and CYP2C8; the human P450s CYP3A3, CYP3A4 and CYP3A5 also catalysed metabolism, but were much less active; and CYP2A6, CYP2C9 and CYP2D6 had little detectable activity. styrene glycol 80-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 8423765-13 1993 Both CYP1A2 and CYP3A4 catalyzed N-dealkylation of propafenone, with specific activities of 0.32 pmol/min/pmol of P450 and 0.16 pmol/min/pmol of P450, respectively. Nitrogen 33-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-118 8423765-13 1993 Both CYP1A2 and CYP3A4 catalyzed N-dealkylation of propafenone, with specific activities of 0.32 pmol/min/pmol of P450 and 0.16 pmol/min/pmol of P450, respectively. Nitrogen 33-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-149 8423765-0 1993 Identification and characterization of the cytochrome P450 enzymes involved in N-dealkylation of propafenone: molecular base for interaction potential and variable disposition of active metabolites. Propafenone 97-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 8423765-13 1993 Both CYP1A2 and CYP3A4 catalyzed N-dealkylation of propafenone, with specific activities of 0.32 pmol/min/pmol of P450 and 0.16 pmol/min/pmol of P450, respectively. Propafenone 51-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-118 8423765-5 1993 We, therefore, characterized the enzyme involved in the formation of N-desalkylpropafenone by using both in vitro [human liver microsomes, specific antibodies or inhibitors, and stably expressed cytochrome P450 (P450) enzymes] and in vivo (formation of N-desalkylpropafenone in patients under conditions of chronic therapy) approaches. N-depropylpropafenone 69-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 206-210 8423765-13 1993 Both CYP1A2 and CYP3A4 catalyzed N-dealkylation of propafenone, with specific activities of 0.32 pmol/min/pmol of P450 and 0.16 pmol/min/pmol of P450, respectively. Propafenone 51-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 145-149 8423765-5 1993 We, therefore, characterized the enzyme involved in the formation of N-desalkylpropafenone by using both in vitro [human liver microsomes, specific antibodies or inhibitors, and stably expressed cytochrome P450 (P450) enzymes] and in vivo (formation of N-desalkylpropafenone in patients under conditions of chronic therapy) approaches. N-depropylpropafenone 69-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 212-216 8423765-5 1993 We, therefore, characterized the enzyme involved in the formation of N-desalkylpropafenone by using both in vitro [human liver microsomes, specific antibodies or inhibitors, and stably expressed cytochrome P450 (P450) enzymes] and in vivo (formation of N-desalkylpropafenone in patients under conditions of chronic therapy) approaches. N-depropylpropafenone 253-274 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 206-210 8423765-5 1993 We, therefore, characterized the enzyme involved in the formation of N-desalkylpropafenone by using both in vitro [human liver microsomes, specific antibodies or inhibitors, and stably expressed cytochrome P450 (P450) enzymes] and in vivo (formation of N-desalkylpropafenone in patients under conditions of chronic therapy) approaches. N-depropylpropafenone 253-274 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 212-216 22217845-0 1992 Inhibitors of P450-dependent steroid biosynthesis: from research to medical treatment. Steroids 29-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 8447150-9 1993 The present results suggest that the P450 enzymes involved in antipyrine metabolism are differentially affected in cirrhosis, but there appear to be no differences in the activity of the enzymes between alcoholic and non-alcoholic cirrhosis. Antipyrine 62-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-41 22217845-1 1992 A number of cytochrome P450-dependent enzymes are major targets for both steroidal and nonsteroidal compounds that may be of use in the treatment of a number of androgen-independent, androgen-, estrogen- and other steroid-dependent diseases. Steroids 73-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-27 22217824-1 1992 In the pathways of steroid hormone biosynthesis there are two major types of enzymes: cytochromes P450 and other steroid oxidoreductases. Steroids 19-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 1598960-2 1992 After the cyclosporine dose was reduced by two thirds, these patients were randomly assigned to treatment with or without ketoconazole, a potent inhibitor of cytochrome P-450, in a double-masked placebo-controlled study. Ketoconazole 122-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-174 1417844-6 1992 Dissociation of P-450 oligomers into monomers was made by addition of 0.2% Triton N-101. Triton N 101 75-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-21 1423839-0 1992 Cytochrome P450 2E1 and 2A6 enzymes as major catalysts for metabolic activation of N-nitrosodialkylamines and tobacco-related nitrosamines in human liver microsomes. n-nitrosodialkylamines 83-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 1423839-0 1992 Cytochrome P450 2E1 and 2A6 enzymes as major catalysts for metabolic activation of N-nitrosodialkylamines and tobacco-related nitrosamines in human liver microsomes. Nitrosamines 126-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-15 1423839-3 1992 The activation of NDMA and NNN by liver microsomes was suggested to be catalyzed more actively by P450 2E1 than by other P450 enzymes because the activities were well correlated with NDMA N-demethylation and aniline p-hydroxylation in different human samples, and purified P450 2E1 had the highest activities in reconstituted monooxygenase systems. aniline 208-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-102 1423839-3 1992 The activation of NDMA and NNN by liver microsomes was suggested to be catalyzed more actively by P450 2E1 than by other P450 enzymes because the activities were well correlated with NDMA N-demethylation and aniline p-hydroxylation in different human samples, and purified P450 2E1 had the highest activities in reconstituted monooxygenase systems. aniline 208-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 1423839-3 1992 The activation of NDMA and NNN by liver microsomes was suggested to be catalyzed more actively by P450 2E1 than by other P450 enzymes because the activities were well correlated with NDMA N-demethylation and aniline p-hydroxylation in different human samples, and purified P450 2E1 had the highest activities in reconstituted monooxygenase systems. aniline 208-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-125 1423839-4 1992 The relatively high contribution of P450 2A6 to the activation of NDEA and NNK was supported by the correlation seen with coumarin 7-hydroxylation in human liver microsomes, and antibodies raised against P450 2A6 inhibited both activities by approximately 50%. coumarin 122-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 1423839-6 1992 Thus, this work indicates that several P450 enzymes, particularly P450 2E1 and 2A6, catalyze metabolic activation of nitrosamine derivatives including N-nitrosodialkylamines and tobacco-smoke-related nitrosamines in human liver microsomes. Nitrosamines 117-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 1423839-6 1992 Thus, this work indicates that several P450 enzymes, particularly P450 2E1 and 2A6, catalyze metabolic activation of nitrosamine derivatives including N-nitrosodialkylamines and tobacco-smoke-related nitrosamines in human liver microsomes. Nitrosamines 117-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 1423839-6 1992 Thus, this work indicates that several P450 enzymes, particularly P450 2E1 and 2A6, catalyze metabolic activation of nitrosamine derivatives including N-nitrosodialkylamines and tobacco-smoke-related nitrosamines in human liver microsomes. n-nitrosodialkylamines 151-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 1423839-6 1992 Thus, this work indicates that several P450 enzymes, particularly P450 2E1 and 2A6, catalyze metabolic activation of nitrosamine derivatives including N-nitrosodialkylamines and tobacco-smoke-related nitrosamines in human liver microsomes. n-nitrosodialkylamines 151-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 1423839-6 1992 Thus, this work indicates that several P450 enzymes, particularly P450 2E1 and 2A6, catalyze metabolic activation of nitrosamine derivatives including N-nitrosodialkylamines and tobacco-smoke-related nitrosamines in human liver microsomes. Nitrosamines 200-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 1423839-6 1992 Thus, this work indicates that several P450 enzymes, particularly P450 2E1 and 2A6, catalyze metabolic activation of nitrosamine derivatives including N-nitrosodialkylamines and tobacco-smoke-related nitrosamines in human liver microsomes. Nitrosamines 200-212 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 1291449-1 1992 Carcinogenic azo dyes (dimethylaminoazobenzene, Sudan I) and N-nitrosamines (N-nitrosomethylaniline, N-nitrosomethylbenzylamine) are oxidized by cytochrome P-450 isoenzymes and peroxidase yielding metabolites which in vitro bind to DNA and transfer RNA (tRNA). p-Dimethylaminoazobenzene 23-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 156-161 1291449-1 1992 Carcinogenic azo dyes (dimethylaminoazobenzene, Sudan I) and N-nitrosamines (N-nitrosomethylaniline, N-nitrosomethylbenzylamine) are oxidized by cytochrome P-450 isoenzymes and peroxidase yielding metabolites which in vitro bind to DNA and transfer RNA (tRNA). n-nitrosamines 61-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 156-161 1291449-1 1992 Carcinogenic azo dyes (dimethylaminoazobenzene, Sudan I) and N-nitrosamines (N-nitrosomethylaniline, N-nitrosomethylbenzylamine) are oxidized by cytochrome P-450 isoenzymes and peroxidase yielding metabolites which in vitro bind to DNA and transfer RNA (tRNA). N-methyl-N-nitrosoaniline 77-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 156-161 1629218-7 1992 The amino acid sequences of six tryptic peptide fragments of cytochrome P-450terp have also been determined. Peptides 40-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-81 1486866-2 1992 Studies with cytochrome P450 (P450) enzymes and glutathione S-transferases are summarized here, and recent work with pyrrolizidine alkaloids, aflatoxins, 4,4"-methylenebis(2-chloroaniline), and ethyl carbamate is discussed. Urethane 194-209 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-34 1306334-3 1992 Different P450 forms are responsible for activation of the various classes of chemical carcinogens including the arylamines, polycyclic aromatic hydrocarbons, nitrosamines and aflatoxins. aniline 113-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-14 1306334-3 1992 Different P450 forms are responsible for activation of the various classes of chemical carcinogens including the arylamines, polycyclic aromatic hydrocarbons, nitrosamines and aflatoxins. Polycyclic Aromatic Hydrocarbons 125-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-14 1306334-3 1992 Different P450 forms are responsible for activation of the various classes of chemical carcinogens including the arylamines, polycyclic aromatic hydrocarbons, nitrosamines and aflatoxins. Nitrosamines 159-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-14 1306334-3 1992 Different P450 forms are responsible for activation of the various classes of chemical carcinogens including the arylamines, polycyclic aromatic hydrocarbons, nitrosamines and aflatoxins. Aflatoxins 176-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-14 1380405-6 1992 Besides the conversion of L-arginine, type I NOS, Ca2+/calmodulin dependently, generates H2O2 and reduces cytochrome c/P450. Arginine 26-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-123 1386614-6 1992 These findings demonstrate that a low dose of spironolactone is effective in the treatment of rosacea in some male patients and suggest that it is possible that changes in the metabolism of sex steroid hormones such as cytochrome p-450 isozymes have some bearing on the etiology of rosacea. Spironolactone 46-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 230-235 1349603-2 1992 Mitochondrial ferredoxins mediate electron transfer from NADPH:ferredoxin oxidoreductase to cytochrome P450 enzymes. NADP 57-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-107 1349603-3 1992 Previous studies on human ferredoxin, in which acidic residues were replaced with neutral amino acids, established that Asp-76 and Asp-79 are are important for binding to both reductase and P450 (Coghlan, V. M., and Vickery, L. E. (1991) J. Biol. Aspartic Acid 120-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 190-194 1349603-3 1992 Previous studies on human ferredoxin, in which acidic residues were replaced with neutral amino acids, established that Asp-76 and Asp-79 are are important for binding to both reductase and P450 (Coghlan, V. M., and Vickery, L. E. (1991) J. Biol. Aspartic Acid 131-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 190-194 1386614-6 1992 These findings demonstrate that a low dose of spironolactone is effective in the treatment of rosacea in some male patients and suggest that it is possible that changes in the metabolism of sex steroid hormones such as cytochrome p-450 isozymes have some bearing on the etiology of rosacea. Steroids 194-210 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 230-235 1316854-0 1992 Regulation of the cholesterol side-chain cleavage cytochrome P-450 and adrenodoxin mRNAs in cultured choriocarcinoma cells. Cholesterol 18-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-82 1588920-4 1992 Human lung microsomes contain approximately 10 pmol of P-450/mg of protein, on the basis of Fe2+.CO versus Fe2+ difference spectra of the eluates obtained from an octylamino-agarose column. ammonium ferrous sulfate 92-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 1588920-4 1992 Human lung microsomes contain approximately 10 pmol of P-450/mg of protein, on the basis of Fe2+.CO versus Fe2+ difference spectra of the eluates obtained from an octylamino-agarose column. Carbon Monoxide 97-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 1588920-4 1992 Human lung microsomes contain approximately 10 pmol of P-450/mg of protein, on the basis of Fe2+.CO versus Fe2+ difference spectra of the eluates obtained from an octylamino-agarose column. ammonium ferrous sulfate 107-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 1588920-4 1992 Human lung microsomes contain approximately 10 pmol of P-450/mg of protein, on the basis of Fe2+.CO versus Fe2+ difference spectra of the eluates obtained from an octylamino-agarose column. Sepharose 174-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 1588920-5 1992 The partially purified P-450 preparations from two human lung microsomal samples showed high activities for the conversion of both (+)- and (-)-isomers of 7,8-dihydroxy-7,8-dihydrobenzo(a)pyrene to genotoxic products. benzo(a)pyrene 7,8-dihydrodiol 155-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-28 1588920-6 1992 After DEAE-cellulose column chromatography, a partially purified P-450 fraction containing polypeptides of Mr 52,000 and 58,000 was obtained from the early fraction of the octylamino-agarose column eluate, and an electrophoretically homogeneous protein having a molecular weight of approximately 52,000 was recovered from a latter fraction. octylazanide 172-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-70 1588920-6 1992 After DEAE-cellulose column chromatography, a partially purified P-450 fraction containing polypeptides of Mr 52,000 and 58,000 was obtained from the early fraction of the octylamino-agarose column eluate, and an electrophoretically homogeneous protein having a molecular weight of approximately 52,000 was recovered from a latter fraction. Sepharose 183-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-70 1588920-8 1992 The protein from the latter octylamino-agarose fraction was immunoreactive with anti-rat P-450 1A2 and anti-human P-450 1A2 but not with antibodies raised against other P-450 enzymes or autoimmune antibodies that specifically recognize human P-450 1A2. Sepharose 39-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-94 1588920-8 1992 The protein from the latter octylamino-agarose fraction was immunoreactive with anti-rat P-450 1A2 and anti-human P-450 1A2 but not with antibodies raised against other P-450 enzymes or autoimmune antibodies that specifically recognize human P-450 1A2. Sepharose 39-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-119 1588920-8 1992 The protein from the latter octylamino-agarose fraction was immunoreactive with anti-rat P-450 1A2 and anti-human P-450 1A2 but not with antibodies raised against other P-450 enzymes or autoimmune antibodies that specifically recognize human P-450 1A2. Sepharose 39-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-119 1588920-8 1992 The protein from the latter octylamino-agarose fraction was immunoreactive with anti-rat P-450 1A2 and anti-human P-450 1A2 but not with antibodies raised against other P-450 enzymes or autoimmune antibodies that specifically recognize human P-450 1A2. Sepharose 39-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-119 1588920-9 1992 A tryptic peptide was isolated from the preparation, and the amino acid sequence matched that of human P-450 1A1 perfectly (residues 31-48) but not that of human P-450 1A2. Peptides 10-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-108 1588920-10 1992 All of nine human lung microsomal samples examined contained proteins that were immunoreactive with rabbit anti-rat P-450 1A2 and catalyzed the activation of 7,8-dihydroxy-7,8-dihydrobenzo(a)pyrene. benzo(a)pyrene 7,8-dihydrodiol 158-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-121 1588920-13 1992 Thus, this work clearly shows that human lung microsomes contain at least two major P-450 enzymes; human P-450 1A1 is present in lungs and can actually catalyze the activation of environmental procarcinogens, including polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 219-251 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-89 1588920-13 1992 Thus, this work clearly shows that human lung microsomes contain at least two major P-450 enzymes; human P-450 1A1 is present in lungs and can actually catalyze the activation of environmental procarcinogens, including polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 219-251 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 105-110 1918043-2 1991 The reactions of cytochromes P450 IA1, IIB1, IIB2, and IIE1 with phenyldiazene yield complexes with absorption maxima at either 474 or 480 nm. phenyldiazene 65-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-43 1540227-0 1992 Ridogrel: a selective inhibitor of the cytochrome P450-dependent thromboxane synthesis. Thromboxanes 65-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 1739747-5 1992 The first 35 NH2-terminal amino acid sequence of P-450HK omega had about 70% homology with those of rabbit kidney fatty acid omega-hydroxylases of the P-450 IVA gene subfamily, P-450ka-1, P-450ka-2, and P-450kd, except for four undetermined residues. Fatty Acids 114-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-54 1794978-3 1991 In the oxidized form, the Soret band was red-shifted as compared with the typical ferric low-spin form of P450 and the beta band was more intense than the alpha band. Ferric enterobactin ion 82-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-110 1794978-4 1991 In the reduced form, two Soret peaks were observable at 447 and 423 nm and their relative heights were dependent on pH, indicating the existence of two interconvertible states of ferrous Lys-mutated P450 which are in equilibrium. Lysine 187-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 199-203 1794978-5 1991 In addition, the interaction of external ligands with the P450 heme iron was profoundly inhibited both in the oxidized and reduced forms. Heme 63-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 1794978-5 1991 In addition, the interaction of external ligands with the P450 heme iron was profoundly inhibited both in the oxidized and reduced forms. Iron 68-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-62 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Nitrogen 42-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-193 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Nitrogen 42-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 233-237 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Lysine 54-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-193 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Lysine 54-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 233-237 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Heme 160-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-193 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Heme 160-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 233-237 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Iron 165-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-193 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Iron 165-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 233-237 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Lysine 177-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-193 1794978-6 1991 These findings suggest that epsilon-amino nitrogen of Lys-301, which was introduced by amino acid substitution, occupies the 6th coordination position with the heme iron of the Lys-mutated P450, because, owing to conformation of the P450 protein, the epsilon-amino group may be located at just the right position for coordination as the internal 6th ligand. Lysine 177-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 233-237 1540227-1 1992 Ridogrel [(E)-5-[[[(3-pyridinyl)[3-(trifluoromethyl)phenyl] methylene]amino]oxy] pentanoic acid] is a potent inhibitor of the P450-dependent human platelet thromboxane A2 (TxA2) synthase. ridogrel 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 1540227-1 1992 Ridogrel [(E)-5-[[[(3-pyridinyl)[3-(trifluoromethyl)phenyl] methylene]amino]oxy] pentanoic acid] is a potent inhibitor of the P450-dependent human platelet thromboxane A2 (TxA2) synthase. (e)-5-[[[(3-pyridinyl)[3-(trifluoromethyl)phenyl] methylene]amino]oxy] pentanoic acid 10-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-130 1540227-6 1992 These results indicate that ridogrel binds stoichiometrically and suggest that inhibition of thromboxane synthesis may originate from liganding of its basic nitrogen to the haem-iron of P450 and from the attachment of the hydrophobic carboxylic side chain to or near the substrate binding place. Thromboxanes 93-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 186-190 1540227-8 1992 At a high concentration (10 microM), ridogrel has a slight, if any, effect on the P450-mediated cholesterol synthesis in human liver and hepatoma cells and androgen synthesis from 17 alpha-hydroxy-20-dihydroprogesterone or pregnenolone in subcellular fractions from rat testes. Cholesterol 96-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 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 2 subfamily B member 6 Homo sapiens 185-194 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 2 subfamily B member 6 Homo sapiens 185-194 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 2 subfamily B member 6 Homo sapiens 185-194 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 2 subfamily B member 6 Homo sapiens 185-194 1770192-5 1991 P-450 induction also explains depletion (and enhanced toxicity) of nutritional factors such as vitamin A. Vitamin A 95-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 1844820-1 1991 A variant CYP2D6(C) P450 protein was found in a liver characterized by deficient microsomal metabolism of bufuralol and sparteine, prototypical substrates for the debrisoquine-sparteine drug oxidation polymorphism. bufuralol 106-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 1844820-1 1991 A variant CYP2D6(C) P450 protein was found in a liver characterized by deficient microsomal metabolism of bufuralol and sparteine, prototypical substrates for the debrisoquine-sparteine drug oxidation polymorphism. Sparteine 120-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 1844820-1 1991 A variant CYP2D6(C) P450 protein was found in a liver characterized by deficient microsomal metabolism of bufuralol and sparteine, prototypical substrates for the debrisoquine-sparteine drug oxidation polymorphism. Debrisoquin 163-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 1844820-1 1991 A variant CYP2D6(C) P450 protein was found in a liver characterized by deficient microsomal metabolism of bufuralol and sparteine, prototypical substrates for the debrisoquine-sparteine drug oxidation polymorphism. Sparteine 176-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-24 1844820-5 1991 The CYP2D6(C) P450, produced in HepG2 cells using vaccinia virus mediated cDNA expression displayed Km values toward bufuralol, debrisoquine and sparteine that were not significantly different from wild type CYP2D6. bufuralol 117-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 1844820-5 1991 The CYP2D6(C) P450, produced in HepG2 cells using vaccinia virus mediated cDNA expression displayed Km values toward bufuralol, debrisoquine and sparteine that were not significantly different from wild type CYP2D6. Debrisoquin 128-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 1844820-5 1991 The CYP2D6(C) P450, produced in HepG2 cells using vaccinia virus mediated cDNA expression displayed Km values toward bufuralol, debrisoquine and sparteine that were not significantly different from wild type CYP2D6. Sparteine 145-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-18 1651809-0 1991 Metabolic activation of 4-ipomeanol by complementary DNA-expressed human cytochromes P-450: evidence for species-specific metabolism. 4-ipomeanol 24-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-90 1651809-1 1991 4-Ipomeanol is a pulmonary toxin in cattle and rodents that is metabolically activated by cytochromes P-450 (P-450s). 4-ipomeanol 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-107 1651809-1 1991 4-Ipomeanol is a pulmonary toxin in cattle and rodents that is metabolically activated by cytochromes P-450 (P-450s). 4-ipomeanol 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 1651809-2 1991 P-450-mediated activation of 4-ipomeanol to DNA binding metabolites was evaluated using a vaccinia virus complementary DNA expression system and an in situ DNA-binding assay. 4-ipomeanol 29-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 1705382-0 1991 Biosynthesis of P450 products of arachidonic acid in humans: increased formation in cardiovascular disease. Arachidonic Acid 33-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 1680658-0 1991 Metabolism of nicotine by rat liver cytochromes P-450. Nicotine 14-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-53 1680658-3 1991 The cytochromes P-450 have long been implicated in the first step in the conversion of nicotine to nicotine delta 1"(5")-iminium ion. Nicotine 87-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-21 1680658-3 1991 The cytochromes P-450 have long been implicated in the first step in the conversion of nicotine to nicotine delta 1"(5")-iminium ion. nicotine delta 1"(5")-iminium 99-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-21 1680658-5 1991 A constitutive form of P-450 is also implicated in nicotine metabolism, while purified P-450IA1 and P-450IIC6 show no detectable activity. Nicotine 51-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-28 1680658-8 1991 Our results support the notion that nicotine metabolism to cotinine by P-450 enzymes is highly species dependent. Nicotine 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-76 1680658-8 1991 Our results support the notion that nicotine metabolism to cotinine by P-450 enzymes is highly species dependent. Cotinine 59-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-76 1680658-9 1991 Thus, it is unwise in some cases to extrapolate results obtained by animal model study to the possible role of specific forms of the P-450 enzymes in nicotine metabolism in humans. Nicotine 150-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-138 16668240-3 1991 Evidence that cytochromes P-450 are involved in the detoxification of herbicides (chlorotoluron, primsulfuron, and diclofop) includes photoreversible CO inhibition of the reactions, and a requirement for O(2) and NADPH. chlortoluron 82-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-31 16668240-3 1991 Evidence that cytochromes P-450 are involved in the detoxification of herbicides (chlorotoluron, primsulfuron, and diclofop) includes photoreversible CO inhibition of the reactions, and a requirement for O(2) and NADPH. primsulfuron 97-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-31 16668240-3 1991 Evidence that cytochromes P-450 are involved in the detoxification of herbicides (chlorotoluron, primsulfuron, and diclofop) includes photoreversible CO inhibition of the reactions, and a requirement for O(2) and NADPH. Diclofop 115-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-31 16668240-3 1991 Evidence that cytochromes P-450 are involved in the detoxification of herbicides (chlorotoluron, primsulfuron, and diclofop) includes photoreversible CO inhibition of the reactions, and a requirement for O(2) and NADPH. o(2) 204-208 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-31 16668240-3 1991 Evidence that cytochromes P-450 are involved in the detoxification of herbicides (chlorotoluron, primsulfuron, and diclofop) includes photoreversible CO inhibition of the reactions, and a requirement for O(2) and NADPH. NADP 213-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-31 16668240-4 1991 Several cytochromes P-450, M(r) 45,000 to 65,000, have been isolated, including hydroxylases of cinnamic acid, 3,9-dihydroxypterocarpan, and digitoxin. cinnamic acid 96-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-25 16668240-4 1991 Several cytochromes P-450, M(r) 45,000 to 65,000, have been isolated, including hydroxylases of cinnamic acid, 3,9-dihydroxypterocarpan, and digitoxin. 3,9-dihydroxypterocarpan 111-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-25 16668240-4 1991 Several cytochromes P-450, M(r) 45,000 to 65,000, have been isolated, including hydroxylases of cinnamic acid, 3,9-dihydroxypterocarpan, and digitoxin. Digitoxin 141-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-25 16668240-7 1991 Immunological techniques and specific inhibitors (triazoles, imidazole derivatives) are being used to characterize plant cytochromes P-450 and the NADPH:cytochrome P-450 reductase. Triazoles 50-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-138 16668240-7 1991 Immunological techniques and specific inhibitors (triazoles, imidazole derivatives) are being used to characterize plant cytochromes P-450 and the NADPH:cytochrome P-450 reductase. imidazole 61-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-138 2051622-0 1991 [Immunohistochemical Study of 3-methylcholanthrene inducible cytochrome P-450 in the stomach and liver--a guide of postoperative chemotherapy in gastric cancer]. Methylcholanthrene 30-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-77 2051622-1 1991 Immunohistochemical determination of 3-methylcholanthrene (MC) inducible cytochrome P-450 (MC-P-450) was investigated in rat and human tissue, and its clinical availability was discussed. Methylcholanthrene 37-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-99 2051622-1 1991 Immunohistochemical determination of 3-methylcholanthrene (MC) inducible cytochrome P-450 (MC-P-450) was investigated in rat and human tissue, and its clinical availability was discussed. Methylcholanthrene 59-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-99 2051622-2 1991 Induced by MC, MC-P-450 in rat liver and stomach was well stained immunohistochemically, showing clear contrast against control without induction. Methylcholanthrene 11-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-23 2007137-10 1991 The aromatase P-450 was subjected to SDS-polyacrylamide gel electrophoresis in increasing quantities. Sodium Dodecyl Sulfate 37-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-19 2007137-10 1991 The aromatase P-450 was subjected to SDS-polyacrylamide gel electrophoresis in increasing quantities. polyacrylamide 41-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-19 1714723-8 1991 The highly conserved cysteine-containing sequence involved in the heme-binding site of P450 was found near the carboxyl terminus (residues 472-492). Cysteine 21-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 1714723-8 1991 The highly conserved cysteine-containing sequence involved in the heme-binding site of P450 was found near the carboxyl terminus (residues 472-492). Heme 66-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 1997003-1 1991 Catalysis by a cytochrome P450 IIC different from that responsible for mephenytoin hydroxylation. Mephenytoin 71-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 1997003-7 1991 However cross inhibition experiments indicate that tienilic acid hydroxylation and mephenytoin hydroxylation, a typical reaction of some human liver P450 IIC isoenzymes, are not catalysed by the same member of the P450 IIC subfamily. Ticrynafen 51-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-153 1997003-7 1991 However cross inhibition experiments indicate that tienilic acid hydroxylation and mephenytoin hydroxylation, a typical reaction of some human liver P450 IIC isoenzymes, are not catalysed by the same member of the P450 IIC subfamily. Ticrynafen 51-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 214-218 1997003-7 1991 However cross inhibition experiments indicate that tienilic acid hydroxylation and mephenytoin hydroxylation, a typical reaction of some human liver P450 IIC isoenzymes, are not catalysed by the same member of the P450 IIC subfamily. Mephenytoin 83-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-153 1997003-7 1991 However cross inhibition experiments indicate that tienilic acid hydroxylation and mephenytoin hydroxylation, a typical reaction of some human liver P450 IIC isoenzymes, are not catalysed by the same member of the P450 IIC subfamily. Mephenytoin 83-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 214-218 1988174-9 1991 In a reconstituted system containing purified forms of human P450, P450IA2 was the most active in catalyzing 3-MeO-AAB, followed by P450IIIA4 and P450MP. 3-methoxy-4-aminoazobenzene 109-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-74 1784247-4 1991 Substitution of basic amino acids at the P450 N terminus or replacing the N terminus of P450 with a secretory signal sequence results in partial or complete translocation across the membrane. Amino Acids, Basic 16-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-45 2241965-4 1990 The antimycotic drug ketoconazole (Ki = 22nM) as well as the isozyme specific P450 inhibitor alpha-naphthoflavone (Ki = 1.2 nM) were shown to be strong inhibitors of human P450IA1. alpha-naphthoflavone 93-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-82 2241188-5 1990 From the results obtained, reasonable consecutive administration of tegafur would be possible by immunohistochemical checking the P450 in the gastric wall. Tegafur 68-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-134 2263067-1 1990 Amphetamine is metabolized by cytochrome P-450 (P450) to p-hydroxyamphetamine and phenylacetone in mammalian species. Amphetamine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-46 2091733-2 1990 Currently, representatives of two classes of EBI antifungals are available: the squalene epoxidase inhibitors and those that interfere with cytochrome P450-dependent ergosterol synthesis. 1,2-Diisothiocyanatoethane 45-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-155 2091733-2 1990 Currently, representatives of two classes of EBI antifungals are available: the squalene epoxidase inhibitors and those that interfere with cytochrome P450-dependent ergosterol synthesis. Ergosterol 166-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-155 2091733-8 1990 All the azole antifungals inhibit the cytochrome P450-dependent, 14 alpha-demethylase, a key enzyme in the synthesis of ergosterol, the main sterol in most fungal cells. Azoles 8-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 2091733-8 1990 All the azole antifungals inhibit the cytochrome P450-dependent, 14 alpha-demethylase, a key enzyme in the synthesis of ergosterol, the main sterol in most fungal cells. Ergosterol 120-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-53 2091733-9 1990 Of all the azoles tested, itraconazole shows the highest affinity for the cytochrome P450 involved. Azoles 11-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 2091733-9 1990 Of all the azoles tested, itraconazole shows the highest affinity for the cytochrome P450 involved. Itraconazole 26-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-89 2091733-11 1990 Itraconazole"s high affinity for the fungal P450 originates from its triazole group as well as from the nonligating lipophilic tail. Itraconazole 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 2091733-11 1990 Itraconazole"s high affinity for the fungal P450 originates from its triazole group as well as from the nonligating lipophilic tail. Triazoles 69-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-48 2263067-1 1990 Amphetamine is metabolized by cytochrome P-450 (P450) to p-hydroxyamphetamine and phenylacetone in mammalian species. Amphetamine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 2263067-1 1990 Amphetamine is metabolized by cytochrome P-450 (P450) to p-hydroxyamphetamine and phenylacetone in mammalian species. p-Hydroxyamphetamine 57-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-46 2263067-1 1990 Amphetamine is metabolized by cytochrome P-450 (P450) to p-hydroxyamphetamine and phenylacetone in mammalian species. p-Hydroxyamphetamine 57-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 2263067-1 1990 Amphetamine is metabolized by cytochrome P-450 (P450) to p-hydroxyamphetamine and phenylacetone in mammalian species. 1-phenyl-2-propanone 82-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-46 2263067-1 1990 Amphetamine is metabolized by cytochrome P-450 (P450) to p-hydroxyamphetamine and phenylacetone in mammalian species. 1-phenyl-2-propanone 82-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-52 2263067-10 1990 These data show that quinidine inhibits in vivo metabolism of amphetamine in rats and suggest that amphetamine metabolism may, in part, be mediated by an isozyme of P450 which displays genetic polymorphism. Quinidine 21-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-169 2263067-10 1990 These data show that quinidine inhibits in vivo metabolism of amphetamine in rats and suggest that amphetamine metabolism may, in part, be mediated by an isozyme of P450 which displays genetic polymorphism. Amphetamine 62-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-169 2263067-10 1990 These data show that quinidine inhibits in vivo metabolism of amphetamine in rats and suggest that amphetamine metabolism may, in part, be mediated by an isozyme of P450 which displays genetic polymorphism. Amphetamine 99-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 165-169 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 2 subfamily B member 6 Homo sapiens 64-69 2397214-2 1990 Spermine, a polycation carrying a charge of 4 +, and potassium, a monovalent cation, were shown to differently cause an increase of high-spin content of camphor-bound cytochrome P-450. Potassium 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-183 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 2 subfamily B member 6 Homo sapiens 64-69 2397214-2 1990 Spermine, a polycation carrying a charge of 4 +, and potassium, a monovalent cation, were shown to differently cause an increase of high-spin content of camphor-bound cytochrome P-450. Camphor 153-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-183 2397214-2 1990 Spermine, a polycation carrying a charge of 4 +, and potassium, a monovalent cation, were shown to differently cause an increase of high-spin content of camphor-bound cytochrome P-450. Spermine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-183 2397214-6 1990 Cytochrome P-420 was produced from cytochrome P-450 by hydrostatic pressure in the presence of potassium, spermine, and cysteine. Potassium 95-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-51 2397214-6 1990 Cytochrome P-420 was produced from cytochrome P-450 by hydrostatic pressure in the presence of potassium, spermine, and cysteine. Spermine 106-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-51 2133086-2 1990 Of the eight compounds tested, gestodene was found to be particularly active as a mechanism-based inactivator of P-450 IIIA4. Gestodene 31-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-118 2397214-6 1990 Cytochrome P-420 was produced from cytochrome P-450 by hydrostatic pressure in the presence of potassium, spermine, and cysteine. Cysteine 120-128 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-51 2133086-8 1990 Consideration of structure/activity relationships among the 17 alpha-acetylenic steroids examined indicates that the delta 15 double bond is critical but is not in itself sufficient for the inactivation process, which is postulated to result from attack of P-450 on the substituted acetylenic carbon and lead to porphyrin N-alkylation. alpha-acetylenic steroids 63-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 257-262 2133086-8 1990 Consideration of structure/activity relationships among the 17 alpha-acetylenic steroids examined indicates that the delta 15 double bond is critical but is not in itself sufficient for the inactivation process, which is postulated to result from attack of P-450 on the substituted acetylenic carbon and lead to porphyrin N-alkylation. Carbon 293-299 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 257-262 2133086-8 1990 Consideration of structure/activity relationships among the 17 alpha-acetylenic steroids examined indicates that the delta 15 double bond is critical but is not in itself sufficient for the inactivation process, which is postulated to result from attack of P-450 on the substituted acetylenic carbon and lead to porphyrin N-alkylation. porphyrin n 312-323 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 257-262 2182263-2 1990 Cytochrome P450 (P450) is the collective term for a group of related enzymes or isozymes which are responsible for the oxidation of numerous drugs and other foreign compounds, as well as many endogenous substrates including prostaglandins, fatty acids and steroids. Prostaglandins 224-238 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 2370296-12 1990 Northern analysis performed on total RNA obtained from forskolin- or hCG-stimulated granulosa-lutein cells confirmed that the increase in aromatase activity was associated with a corresponding increase in levels of mRNA specific for aromatase cytochrome P-450. Colforsin 55-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 254-259 2370296-13 1990 Levels of mRNA encoding cholesterol side-chain cleavage cytochrome P-450 were similarly increased in cells treated with forskolin compared with unstimulated values at each of the time points investigated. Cholesterol 24-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-72 2370296-13 1990 Levels of mRNA encoding cholesterol side-chain cleavage cytochrome P-450 were similarly increased in cells treated with forskolin compared with unstimulated values at each of the time points investigated. Colforsin 120-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-72 2337348-0 1990 Selective detection of mRNA forms encoding the major phenobarbital inducible cytochromes P450 and other members of the P450IIB family by the RNAse A protection assay. Phenobarbital 53-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-93 2353556-3 1990 Both pyridoglutethimide and aminoglutethimide inhibited the aromatase activity of uterine leiomyoma and cultured choriocarcinoma Enami cells as well as immunopurified human placental aromatase cytochrome P-450 by more than 90%, with IC50 values of 10-19 and 5-7 mumol/l, respectively. rogletimide 5-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 204-209 2353556-3 1990 Both pyridoglutethimide and aminoglutethimide inhibited the aromatase activity of uterine leiomyoma and cultured choriocarcinoma Enami cells as well as immunopurified human placental aromatase cytochrome P-450 by more than 90%, with IC50 values of 10-19 and 5-7 mumol/l, respectively. Aminoglutethimide 28-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 204-209 2180561-0 1990 Mutagenic activation of 2-amino-3-methylimidazo[4,5-f]quinoline by complementary DNA-expressed human liver P-450. 2-amino-3-methylimidazo(4,5-f)quinoline 24-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-112 2180561-2 1990 Using the Ames test, each expressed P-450 was examined for its ability to activate to mutagenic products the compounds, 2-amino-3-methylimidazo[4,5-f]quinoline, 2-amino-3,4-dimethylimidazo[4,5-f]quinoline, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline, and 2-amino-3,4,8-trimethylimidazo[4,5-f]quinoxaline, respectively. 2-amino-3-methylimidazo(4,5-f)quinoline 120-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-41 2180561-2 1990 Using the Ames test, each expressed P-450 was examined for its ability to activate to mutagenic products the compounds, 2-amino-3-methylimidazo[4,5-f]quinoline, 2-amino-3,4-dimethylimidazo[4,5-f]quinoline, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline, and 2-amino-3,4,8-trimethylimidazo[4,5-f]quinoxaline, respectively. 2-amino-3,4-dimethylimidazo(4,5-f)quinoline 161-204 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-41 2180561-2 1990 Using the Ames test, each expressed P-450 was examined for its ability to activate to mutagenic products the compounds, 2-amino-3-methylimidazo[4,5-f]quinoline, 2-amino-3,4-dimethylimidazo[4,5-f]quinoline, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline, and 2-amino-3,4,8-trimethylimidazo[4,5-f]quinoxaline, respectively. 2-amino-3,8-dimethylimidazo(4,5-f)quinoxaline 206-251 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-41 2180561-2 1990 Using the Ames test, each expressed P-450 was examined for its ability to activate to mutagenic products the compounds, 2-amino-3-methylimidazo[4,5-f]quinoline, 2-amino-3,4-dimethylimidazo[4,5-f]quinoline, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline, and 2-amino-3,4,8-trimethylimidazo[4,5-f]quinoxaline, respectively. 3,4,8-trimethylimidazo(4,5-f)quinoxalin-2-amine 257-305 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-41 2180561-3 1990 Three forms of human P-450 significantly activated 2-amino-3-methylimidazo[4,5-f]quinoline when the latter was at high substrate concentrations, but only a single form, P-450IA2, showed very high activation of all promutagens at lower substrate concentrations. 2-amino-3-methylimidazo(4,5-f)quinoline 51-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-26 2162057-0 1990 Five of 12 forms of vaccinia virus-expressed human hepatic cytochrome P450 metabolically activate aflatoxin B1. Aflatoxin B1 98-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-74 2162057-4 1990 The same P450s catalyzed conversion of aflatoxin B1 to DNA-bound derivatives as judged by an in situ assay in which the radiolabeled carcinogen was incubated with cells expressing the individual P450 forms. Aflatoxin B1 39-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-13 2162057-8 1990 These results establish that metabolic activation of aflatoxin B1 in human liver involves the contribution of multiple forms of P450. Aflatoxin B1 53-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-132 2318818-0 1990 Mutagenesis of a single hydrogen bond in cytochrome P-450 alters cation binding and heme solvation. Hydrogen 24-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-57 2318818-0 1990 Mutagenesis of a single hydrogen bond in cytochrome P-450 alters cation binding and heme solvation. Heme 84-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-57 2326279-0 1990 Substrate-, hormone-, and cAMP-regulated cytochrome P450 degradation. Cyclic AMP 26-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-56 2182263-2 1990 Cytochrome P450 (P450) is the collective term for a group of related enzymes or isozymes which are responsible for the oxidation of numerous drugs and other foreign compounds, as well as many endogenous substrates including prostaglandins, fatty acids and steroids. Fatty Acids 240-251 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 2182263-2 1990 Cytochrome P450 (P450) is the collective term for a group of related enzymes or isozymes which are responsible for the oxidation of numerous drugs and other foreign compounds, as well as many endogenous substrates including prostaglandins, fatty acids and steroids. Steroids 256-264 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-22 2306418-3 1990 This finding suggests that 3-hydroxylation, in part, is catalyzed by the same isoenzyme of cytochrome P450, P450db1 which oxidizes sparteine. Sparteine 131-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-106 2306418-4 1990 Otherwise, no significant phenotypic differences in total or metabolic clearance were found and it is concluded that the metabolism of quinidine is largely carried out by P450 isoenzymes different from P450db1. Quinidine 135-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-175 2400826-1 1990 The adaptive response of the liver to phenobarbital is characterized by a strong cell hypertrophy and coordinate induction of specific P450 forms (IIB1, 2; IIC7, IIIA1). Phenobarbital 38-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-151 2198032-1 1990 Ethanol has been shown to have a multitude of acute and chronic interactions with xenobiotic agents, many of which can now be explained on the basis of the existence of a newly recognized microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450 (P450IIE1). Ethanol 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 263-268 2198032-1 1990 Ethanol has been shown to have a multitude of acute and chronic interactions with xenobiotic agents, many of which can now be explained on the basis of the existence of a newly recognized microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450 (P450IIE1). Ethanol 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 270-278 2198032-1 1990 Ethanol has been shown to have a multitude of acute and chronic interactions with xenobiotic agents, many of which can now be explained on the basis of the existence of a newly recognized microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450 (P450IIE1). Ethanol 199-206 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 263-268 2198032-1 1990 Ethanol has been shown to have a multitude of acute and chronic interactions with xenobiotic agents, many of which can now be explained on the basis of the existence of a newly recognized microsomal ethanol oxidizing system (MEOS) involving a specific cytochrome P-450 (P450IIE1). Ethanol 199-206 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 270-278 2198032-6 1990 Most importantly, the alcohol-inducible form (P450IIE1) has a unique capacity to activate xenobiotic agents to toxic metabolites, thereby explaining the unusual susceptibility of the alcoholic to the adverse effects of other drugs, hepatotoxic agents, carcinogens and even vitamins. Alcohols 22-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-54 2192911-4 1990 Xenobiotics may lead to liver injury after biotransformation to highly reactive electrophilic metabolites (mainly cytochrome P-450 mediated), which easily conjugate with GSH, thus producing GSH depletion. Glutathione 170-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-130 2183970-3 1990 Some of the moieties that we generally accept as intermediates (i.e., high-valent iron oxygen complex in cytochrome P-450 reactions) would be extremely hard to characterize were it not for biomimetic models and more stable analogs such as peroxidase Compound I complexes. iron oxygen 82-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-121 2192911-4 1990 Xenobiotics may lead to liver injury after biotransformation to highly reactive electrophilic metabolites (mainly cytochrome P-450 mediated), which easily conjugate with GSH, thus producing GSH depletion. Glutathione 190-193 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-130 2134689-2 1990 Microsomal 4"-hydroxylation of S-mephenytoin, but not of R-mephenytoin, was correlated to the content of cytochrome P-450 human-2. Mephenytoin 31-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-121 2122480-0 1990 Studies on the mechanism of monoclonal antibody inhibition of enzyme activity of phenobarbital-induced cytochrome P-450. Phenobarbital 81-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-119 2122480-1 1990 Four monoclonal antibodies (MAbs) to phenobarbital-induced cytochrome P-450 (PB-P-450) show different patterns of inhibition of PB-P-450 catalyzed aryl hydrocarbon hydroxylase (AHH), 7-ethoxycoumarin deethylase, benzphetamine demethylase and ethylmorphine demethylase. Phenobarbital 37-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-75 2122480-1 1990 Four monoclonal antibodies (MAbs) to phenobarbital-induced cytochrome P-450 (PB-P-450) show different patterns of inhibition of PB-P-450 catalyzed aryl hydrocarbon hydroxylase (AHH), 7-ethoxycoumarin deethylase, benzphetamine demethylase and ethylmorphine demethylase. Phenobarbital 37-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-85 2122480-1 1990 Four monoclonal antibodies (MAbs) to phenobarbital-induced cytochrome P-450 (PB-P-450) show different patterns of inhibition of PB-P-450 catalyzed aryl hydrocarbon hydroxylase (AHH), 7-ethoxycoumarin deethylase, benzphetamine demethylase and ethylmorphine demethylase. Phenobarbital 37-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-136 2122480-4 1990 The same cytochrome P-450 bound to carbon monoxide, however, can be reduced chemically by sodium dithionite in the presence of the monoclonal antibody. Carbon Monoxide 35-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-25 2134689-4 1990 The expressed P-450 showed stereoselectivities similar to those in human livers for hydroxylations of mephenytoin and hexobarbital enantiomers. Mephenytoin 102-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-19 2122480-4 1990 The same cytochrome P-450 bound to carbon monoxide, however, can be reduced chemically by sodium dithionite in the presence of the monoclonal antibody. Dithionite 90-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 20-25 2134689-5 1990 The P-450 catalyzed benzo[a]pyrene and tolbutamide hydroxylations and cyclophosphamide oxidation, but showed low activity for the mutagenic activation of IQ (2-amino-3-methylimidazo [4, 5-f]quinoline). Benzo(a)pyrene 20-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-9 2134689-5 1990 The P-450 catalyzed benzo[a]pyrene and tolbutamide hydroxylations and cyclophosphamide oxidation, but showed low activity for the mutagenic activation of IQ (2-amino-3-methylimidazo [4, 5-f]quinoline). Tolbutamide 39-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-9 2134689-5 1990 The P-450 catalyzed benzo[a]pyrene and tolbutamide hydroxylations and cyclophosphamide oxidation, but showed low activity for the mutagenic activation of IQ (2-amino-3-methylimidazo [4, 5-f]quinoline). Cyclophosphamide 70-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-9 2134689-5 1990 The P-450 catalyzed benzo[a]pyrene and tolbutamide hydroxylations and cyclophosphamide oxidation, but showed low activity for the mutagenic activation of IQ (2-amino-3-methylimidazo [4, 5-f]quinoline). 2-amino-3-methylimidazo(4,5-f)quinoline 158-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-9 33802239-6 2021 We found that lignans with one or two methylenedioxyphenyl groups inhibited CYP2B6, CYP2C8, CYP2C9, CYP2C19, and CYP2E1 activities in a time- and concentration-dependent like their CYP3A inhibition. Lignans 14-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 33822485-2 2021 Ivosidenib is an inducer of the CYP2B6, CYP2C8, CYP2C9, and CYP3A4 and an inhibitor of P-glycoprotein (P-gp), organic anion transporting polypeptide-1B1/1B3 (OATP1B1/1B3), and organic anion transporter-3 (OAT3) in vitro. ivosidenib 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 33822485-7 2021 The model was also used to predict the DDIs of ivosidenib with CYP2B6, CYP2C8, CYP2C9, P-gp, OATP1B1/1B3, and OAT3 substrates. ivosidenib 47-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 33816071-3 2020 The aim of our study was to investigate the influence of the cytochrome P450 2B6 isozyme CYP2B6 (rs3745274), gamma-aminobutyric acid type A (GABAA) receptor alpha1 subunit GABRA1 (rs2279020) and ATP-binding cassette subfamily B member 1 ABCB1 (rs1045642) gene polymorphisms on propofol therapeutic outcomes in the patients undergoing abdominal hysterectomy. Propofol 277-285 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-80 33816071-3 2020 The aim of our study was to investigate the influence of the cytochrome P450 2B6 isozyme CYP2B6 (rs3745274), gamma-aminobutyric acid type A (GABAA) receptor alpha1 subunit GABRA1 (rs2279020) and ATP-binding cassette subfamily B member 1 ABCB1 (rs1045642) gene polymorphisms on propofol therapeutic outcomes in the patients undergoing abdominal hysterectomy. Propofol 277-285 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-95 33233444-4 2020 CPA, an integral component of CHOP, is a prodrug that requires CYP2B6-mediated bioactivation to 4-hydroxy-CPA (4-OH-CPA). Cyclophosphamide 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 33233444-9 2020 CITCO robustly induced the expression of cyp2b10 (murine ortholog of CYP2B6) through hCAR activation and increased plasma concentrations of 4-OH-CPA. 4-oh-cpa 140-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 33806634-0 2021 Physiologically Based Pharmacokinetic Modeling of Bupropion and Its Metabolites in a CYP2B6 Drug-Drug-Gene Interaction Network. Bupropion 50-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 33806634-1 2021 The noradrenaline and dopamine reuptake inhibitor bupropion is metabolized by CYP2B6 and recommended by the FDA as the only sensitive substrate for clinical CYP2B6 drug-drug interaction (DDI) studies. Norepinephrine 4-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 33806634-1 2021 The noradrenaline and dopamine reuptake inhibitor bupropion is metabolized by CYP2B6 and recommended by the FDA as the only sensitive substrate for clinical CYP2B6 drug-drug interaction (DDI) studies. Norepinephrine 4-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 33806634-1 2021 The noradrenaline and dopamine reuptake inhibitor bupropion is metabolized by CYP2B6 and recommended by the FDA as the only sensitive substrate for clinical CYP2B6 drug-drug interaction (DDI) studies. Dopamine 22-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 33806634-1 2021 The noradrenaline and dopamine reuptake inhibitor bupropion is metabolized by CYP2B6 and recommended by the FDA as the only sensitive substrate for clinical CYP2B6 drug-drug interaction (DDI) studies. Dopamine 22-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 33806634-1 2021 The noradrenaline and dopamine reuptake inhibitor bupropion is metabolized by CYP2B6 and recommended by the FDA as the only sensitive substrate for clinical CYP2B6 drug-drug interaction (DDI) studies. Bupropion 50-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-84 33806634-1 2021 The noradrenaline and dopamine reuptake inhibitor bupropion is metabolized by CYP2B6 and recommended by the FDA as the only sensitive substrate for clinical CYP2B6 drug-drug interaction (DDI) studies. Bupropion 50-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 33806634-4 2021 It incorporates CYP2B6-mediated hydroxylation of bupropion, metabolism via CYP2C19 and 11beta-HSD, as well as binding to pharmacological targets. Bupropion 49-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 33806634-6 2021 DDI model performance was evaluated by prediction of clinical studies with rifampicin (CYP2B6 and CYP2C19 inducer), fluvoxamine (CYP2C19 inhibitor) and voriconazole (CYP2B6 and CYP2C19 inhibitor). Rifampin 75-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 33233444-4 2020 CPA, an integral component of CHOP, is a prodrug that requires CYP2B6-mediated bioactivation to 4-hydroxy-CPA (4-OH-CPA). 4-hydroxycyclophosphamide 96-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 33233444-4 2020 CPA, an integral component of CHOP, is a prodrug that requires CYP2B6-mediated bioactivation to 4-hydroxy-CPA (4-OH-CPA). 4-oh-cpa 111-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 33233444-6 2020 We have previously demonstrated that the induction of hepatic CYP2B6 by CITCO, a selective human CAR (hCAR) agonist, results in CHOP"s enhanced antineoplastic effects in vitro. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 72-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 25363652-8 2015 Rilpivirine, etravirine, and efavirenz triggered nuclear translocation of hCAR-WT and increased hCAR target gene (CYP2B6) expression. Rilpivirine 0-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 31674039-5 2019 In this study, dasotraline was evaluated as a selective inactivator of CYP2B6 under reaction phenotyping conditions with human hepatocytes. 4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-amine 15-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 31674039-6 2019 The results show that dasotraline is a very selective inactivator for CYP2B6 with minimal inhibition to other enzymes. 4-(3,4-dichlorophenyl)-1,2,3,4-tetrahydronaphthalen-1-amine 22-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 25363652-8 2015 Rilpivirine, etravirine, and efavirenz triggered nuclear translocation of hCAR-WT and increased hCAR target gene (CYP2B6) expression. etravirine 13-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 34986431-7 2022 When evaluated separately, individual genes (for CYP2C19 and CYP2B6) and the combinatorial algorithm were significant predictors of sertraline blood levels. Sertraline 132-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 34752647-0 2022 Association of CYP2B6 genetic polymorphisms with bupropion and hydroxybupropion exposure: A systematic review and meta-analysis. Bupropion 49-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 34910929-4 2022 The results suggested that vicagrel inhibited CYP2B6 and CYP2C19 potently with apparent IC50 values of 1.6 and 2.0 muM, respectively. methyl 2-(2-acetoxy-6,7-dihydrothieno(3,2-c)pyridin-5(4H)-yl)-2-(2-chlorophenyl)acetate 27-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 34910929-5 2022 In terms of mode of reversible inhibition, vicagrel exhibited mixed-type inhibition of CYP2B6-catalyzed bupropion hydroxylation and noncompetitive inhibition of CYP2C19-mediated S-mephenytoin 4"-hydroxylation with Ki values of 0.19 muM and 1.2 muM, respectively. methyl 2-(2-acetoxy-6,7-dihydrothieno(3,2-c)pyridin-5(4H)-yl)-2-(2-chlorophenyl)acetate 43-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 34910929-5 2022 In terms of mode of reversible inhibition, vicagrel exhibited mixed-type inhibition of CYP2B6-catalyzed bupropion hydroxylation and noncompetitive inhibition of CYP2C19-mediated S-mephenytoin 4"-hydroxylation with Ki values of 0.19 muM and 1.2 muM, respectively. Bupropion 104-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 34910929-6 2022 Vicagrel displayed profound time-dependent inhibition towards CYP2B6 with maximal rate constant of inactivation (kinact) and half-maximal inactivator concentration (KI) values of 0.062 min-1 and 1.52 muM, respectively. methyl 2-(2-acetoxy-6,7-dihydrothieno(3,2-c)pyridin-5(4H)-yl)-2-(2-chlorophenyl)acetate 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 34910929-12 2022 Our study provides inhibitory constants for future DDI prediction between vicagrel and drug substrates of CYP2B6, CYP2C19 and UGT1A6. methyl 2-(2-acetoxy-6,7-dihydrothieno(3,2-c)pyridin-5(4H)-yl)-2-(2-chlorophenyl)acetate 74-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 34752647-12 2022 The findings of this study suggest opportunities to further study precision dosing strategies for bupropion therapy based on CYP2B6 genotype. Bupropion 98-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 34752647-0 2022 Association of CYP2B6 genetic polymorphisms with bupropion and hydroxybupropion exposure: A systematic review and meta-analysis. hydroxybupropion 63-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 34752647-1 2022 INTRODUCTION: Bupropion is metabolized to its active metabolite, hydroxybupropion (HB), by the genetically polymorphic cytochrome P450 2B6 (CYP2B6) enzyme. Bupropion 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-138 34752647-1 2022 INTRODUCTION: Bupropion is metabolized to its active metabolite, hydroxybupropion (HB), by the genetically polymorphic cytochrome P450 2B6 (CYP2B6) enzyme. Bupropion 14-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 34752647-1 2022 INTRODUCTION: Bupropion is metabolized to its active metabolite, hydroxybupropion (HB), by the genetically polymorphic cytochrome P450 2B6 (CYP2B6) enzyme. hydroxybupropion 65-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-138 34752647-1 2022 INTRODUCTION: Bupropion is metabolized to its active metabolite, hydroxybupropion (HB), by the genetically polymorphic cytochrome P450 2B6 (CYP2B6) enzyme. hydroxybupropion 65-81 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 34752647-3 2022 OBJECTIVES: A systematic review and meta-analysis was conducted to quantify the association of bupropion and HB exposure with CYP2B6 variant alleles and genotype-defined metabolizer phenotypes. Bupropion 95-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 34752647-6 2022 The ratio of means (RoM) between CYP2B6 genotype or genotype-defined phenotype groups for bupropion exposure was calculated for each study and combined in a meta-analysis. Bupropion 90-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 34752647-9 2022 The AUCs of HB and the active moiety (bupropion + HB) were significantly reduced in CYP2B6*6 carriers compared to the non-carriers (HB: RoM 0.77, 95% CI 0.71-0.83; active moiety: RoM 0.81, 95% CI 0.75-0.88). Bupropion 38-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 34910759-1 2021 Genetic variants in the OPRM1 and CYP2B6 genes, respectively coding for an opioid receptor and methadone metabolizers, have been linked to negative treatment outcomes in patients undergoing methadone maintenance treatment, with little consensus on their effect. Methadone 95-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 34910759-0 2021 Implications of OPRM1 and CYP2B6 variants on treatment outcomes in methadone-maintained patients in Ontario: Exploring sex differences. Methadone 67-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 34910759-1 2021 Genetic variants in the OPRM1 and CYP2B6 genes, respectively coding for an opioid receptor and methadone metabolizers, have been linked to negative treatment outcomes in patients undergoing methadone maintenance treatment, with little consensus on their effect. Methadone 190-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 34910759-2 2021 This study aims to test the associations between pre-selected SNPs of OPRM1 and CYP2B6 and outcomes of continued opioid use, relapse, and methadone dose. Methadone 138-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 34474119-0 2021 Effect of Albumin and CYP2B6 Polymorphisms on Exposure of Efavirenz: A Population Pharmacokinetic Analysis in Chinese HIV-infected Adults. efavirenz 58-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 34873089-0 2022 Impact of CYP2B6 genotype, TB therapy and formulation on efavirenz pharmacokinetics in infants and children under 40 months of age. efavirenz 57-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 34945777-9 2021 On multivariate analysis, CYP2B6*6 and ABCB1c.3435 C > T genotypes and low pre-treatment low-density lipoprotein (LDL) were significantly associated with higher plasma efavirenz concentration regardless of treatment duration. efavirenz 168-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 34945777-11 2021 (4) Conclusion: Pre-treatment LDL cholesterol and CYP2B6*6 and ABCB1c.3435 C > T genotypes predict efavirenz plasma exposure among HIV-infected children, but treatment-duration-dependent changes in plasma efavirenz exposure due to auto-induction are not statistically significant. efavirenz 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-56 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 2 subfamily B member 6 Homo sapiens 290-296 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 2 subfamily B member 6 Homo sapiens 290-296 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 2 subfamily B member 6 Homo sapiens 290-296 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 2 subfamily B member 6 Homo sapiens 141-147 34474119-9 2021 Efavirenz clearance was significantly influenced by CYP2B6 variants, including rs2099361, rs3745274, and rs2279343, along with albumin and weight. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 34474119-11 2021 Based on the CYP2B6 polymorphisms of patients, the recommended daily doses of efavirenz were 100 mg for CYP2B6 slow metabolizers, 400 or 600 mg for intermediate metabolizers, and 800 or 1000 mg for extensive metabolizers. efavirenz 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 34474119-11 2021 Based on the CYP2B6 polymorphisms of patients, the recommended daily doses of efavirenz were 100 mg for CYP2B6 slow metabolizers, 400 or 600 mg for intermediate metabolizers, and 800 or 1000 mg for extensive metabolizers. efavirenz 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-110 34474119-12 2021 CONCLUSIONS: Polymorphisms of CYP2B6, along with albumin and weight, resulted as the predictors of efavirenz pharmacokinetic variability, which could be used in prescribing optimal efavirenz doses. efavirenz 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 34474119-12 2021 CONCLUSIONS: Polymorphisms of CYP2B6, along with albumin and weight, resulted as the predictors of efavirenz pharmacokinetic variability, which could be used in prescribing optimal efavirenz doses. efavirenz 181-190 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 34721047-11 2021 In contrast, ticagrelor-O-glucuronide weakly inhibited CYP2B6, CYP2C9 and CYP2C19 activity with apparent IC50 values of 45.0, 20.0 and 18.8 muM, respectively. ticagrelor-o-glucuronide 13-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-61 34882770-2 2021 Efavirenz is primarily metabolized by cytochrome P450 2B6 (CYP2B6), with CYP2A6 as an accessory pathway. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-57 34882770-2 2021 Efavirenz is primarily metabolized by cytochrome P450 2B6 (CYP2B6), with CYP2A6 as an accessory pathway. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 34415664-0 2021 Large variability in plasma efavirenz concentration in Papua New Guinea HIV/AIDS patients associated with high frequency of CYP2B6 516T allele. efavirenz 28-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-130 34779706-3 2021 The aims of this study were to determine the cytochrome P450 isoforms responsible for pirfenidone 5-hydroxylation and to evaluate their contributions in human liver microsomes (HLM).Among the recombinant P450 isoforms, CYP1A2, CYP2D6, CYP2C19, CYP2A6, and CYP2B6 were shown to catalyze the 5-hydroxylation of pirfenidone. pirfenidone 309-320 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 256-262 34791718-2 2022 BACKGROUND AND AIMS: CYP2B6, a genetically variable enzyme, converts bupropion to its active metabolite hydroxybupropion. Bupropion 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 34791718-2 2022 BACKGROUND AND AIMS: CYP2B6, a genetically variable enzyme, converts bupropion to its active metabolite hydroxybupropion. hydroxybupropion 104-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 34791718-4 2022 The aim of this study was to determine whether genetically normal (vs. reduced) CYP2B6 activity increases bupropion-aided cessation in African American smokers via higher hydroxybupropion concentration, and whether this differs by sex. Bupropion 106-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 34791718-4 2022 The aim of this study was to determine whether genetically normal (vs. reduced) CYP2B6 activity increases bupropion-aided cessation in African American smokers via higher hydroxybupropion concentration, and whether this differs by sex. hydroxybupropion 171-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 34791718-10 2022 FINDINGS: Normal (vs. reduced) CYP2B6 activity was associated with increased cessation at week 7, which was mediated by higher hydroxybupropion concentration (odds ratio (OR)=1.25, 95% confidence interval (CI)=1.03, 1.78); this mediation effect persisted at week 26 (OR=1.23, 95% CI=1.02, 1.70). hydroxybupropion 127-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 34791718-13 2022 CONCLUSIONS: In African American light-smokers with verified early bupropion use, genetically normal CYP2B6 activity appears to be indirectly associated with greater smoking cessation success in a relationship mediated by higher hydroxybupropion concentration. Bupropion 67-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 34328253-15 2021 Unbound dolutegravir >= 431 nM induced expression of CYP3A4 (>= two-fold) in a dose-dependent manner, while 1.44 muM of unbound dolutegravir induced CYP2B6 expression 2.2 +- 0.3-fold (P = 0.0004). dolutegravir 8-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 34328253-15 2021 Unbound dolutegravir >= 431 nM induced expression of CYP3A4 (>= two-fold) in a dose-dependent manner, while 1.44 muM of unbound dolutegravir induced CYP2B6 expression 2.2 +- 0.3-fold (P = 0.0004). dolutegravir 128-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 34746455-0 2021 The correlation between the level of 3-hydroxypropyl mercapturic acid, CYP2B6 polymorphisms, and hematuria occurrences after cyclophosphamide administration and its bioanalytical methods: A systematic review. Cyclophosphamide 125-141 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-77 34746455-1 2021 Background: Cyclophosphamide (CPA) is a cytotoxic prodrug that needs to be metabolized by cytochrome P450 enzymes, like CYP2B6. Cyclophosphamide 12-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 34746455-1 2021 Background: Cyclophosphamide (CPA) is a cytotoxic prodrug that needs to be metabolized by cytochrome P450 enzymes, like CYP2B6. Cyclophosphamide 30-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-126 34746455-2 2021 Unfortunately, CYP2B6 is a very polymorphic enzyme and can cause a change in 3-hydroxypropyl mercapturic acid (3-HPMA), the most found CYP metabolite in urine levels. S-(3-hydroxypropyl)cysteine N-acetate 77-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 34746455-2 2021 Unfortunately, CYP2B6 is a very polymorphic enzyme and can cause a change in 3-hydroxypropyl mercapturic acid (3-HPMA), the most found CYP metabolite in urine levels. S-(3-hydroxypropyl)cysteine N-acetate 111-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 34746455-4 2021 Also, this review was written to examine the relationship between levels of 3-HPMA in urine, polymorphisms of CYP2B6 enzymes, and the incidence of hematuria after cyclophosphamide administration in cancer patients. Cyclophosphamide 163-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-116 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. S-(3-hydroxypropyl)cysteine N-acetate 38-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. S-(3-hydroxypropyl)cysteine N-acetate 38-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. Cyclophosphamide 136-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. Cyclophosphamide 136-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. Cyclophosphamide 136-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. Cyclophosphamide 221-237 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. Cyclophosphamide 221-237 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. Cyclophosphamide 221-237 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. Acrolein 290-298 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 34115651-9 2021 CYP2B6 516G > T, (P < 0.001) and CYP2B6 983T > C (P = 0.001) were each associated with hair efavirenz concentrations. efavirenz 92-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. Acrolein 290-298 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 34115651-9 2021 CYP2B6 516G > T, (P < 0.001) and CYP2B6 983T > C (P = 0.001) were each associated with hair efavirenz concentrations. efavirenz 92-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. Acrolein 290-298 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 34115651-13 2021 CONCLUSION: This study demonstrated approximately 3-fold and 2-fold higher efavirenz plasma and hair concentrations, respectively, among CYP2B6 516TT compared to 516GG. efavirenz 75-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. S-(3-hydroxypropyl)cysteine N-acetate 303-309 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 173-179 34746455-18 2021 Also, there is a relationship between 3-HPMA levels, CYP2B6 polymorphisms, and the occurrences of hematuria after the administration of cyclophosphamide, which is a type of CYP2B6 polymorph, namely CYP2B6*6, can increase cyclophosphamide hydroxylation so that it can increase the levels of acrolein and 3-HPMA, as its metabolites, and risk of hematuria. S-(3-hydroxypropyl)cysteine N-acetate 303-309 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 198-204 34683865-7 2021 Moreover, CYP3A5 and CYP2B6 phenotypes were related to quetiapine exposure variability, which confirms (for CYP3A5) and suggests (for CYP2B6) that these enzymes play an important role in quetiapine"s metabolism. Quetiapine Fumarate 55-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 34482033-12 2021 The CYP 2B6*4 variant decreased S-methadone CL/F by 18%. Methadone 32-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-11 34482033-12 2021 The CYP 2B6*4 variant decreased S-methadone CL/F by 18%. Fluorine 47-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-11 34329567-2 2021 We evaluated the in vitro drug - drug interaction (DDI) potential of cetagliptin, as well as the pharmacokinetics of cetagliptin and metformin and the interaction between cetagliptin and metformin.Cetagliptin did not inhibit CYP1A2, CYP2C8, CYP2B6, CYP2C9, CYP2C19 and CYP3A4, only has a moderate inhibitory effect on CYP2D6, and did not induce CYP1A2, CYP2B6 and CYP3A4. cetagliptin 197-208 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 353-359 34683865-7 2021 Moreover, CYP3A5 and CYP2B6 phenotypes were related to quetiapine exposure variability, which confirms (for CYP3A5) and suggests (for CYP2B6) that these enzymes play an important role in quetiapine"s metabolism. Quetiapine Fumarate 187-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 34683865-7 2021 Moreover, CYP3A5 and CYP2B6 phenotypes were related to quetiapine exposure variability, which confirms (for CYP3A5) and suggests (for CYP2B6) that these enzymes play an important role in quetiapine"s metabolism. Quetiapine Fumarate 187-197 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-140 34575495-3 2021 The inhibitions of CYP1A2, CYP2B6, and CYP2C19 by resveratrol were stereo-nonselective. Resveratrol 50-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 34397877-3 2021 Present study performs meta-analysis to evaluate the association between CYP2B6 c.516G>T variant and AL risk. Aluminum 101-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 34114066-7 2021 RESULTS: We found a significant linear gene-dose relationship with CYP2B6 coding SNP and formation of 4-hydroxycyclophosphamide. 4-hydroxycyclophosphamide 102-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 34165800-3 2021 Candidate gene studies of smoking phenotypes have identified several pharmacogenes implicated in nicotine"s pharmacokinetics (CYP2A6, CYP2B6, CYP2A13, FMOs, UGTs, and OCT2), and nicotine"s pharmacodynamic response in the central nervous system (nicotinic acetylcholine receptors, as well as through the dopaminergic and serotonergic systems). Nicotine 97-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-140 34397877-8 2021 Our results indicated that CYP2B6 c.516G>T variant was significantly related to an increased the risk of AL under dominant model, recessive model, homozygote model, and allelic model. Aluminum 105-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 34397877-11 2021 CONCLUSION: Our meta-analysis indicated that the CYP2B6 variant is significantly associated with AL risk, in which CYP2B6 c.516G>T is related to an increased risk of AL. Aluminum 97-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 34397877-11 2021 CONCLUSION: Our meta-analysis indicated that the CYP2B6 variant is significantly associated with AL risk, in which CYP2B6 c.516G>T is related to an increased risk of AL. Aluminum 97-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 34397877-11 2021 CONCLUSION: Our meta-analysis indicated that the CYP2B6 variant is significantly associated with AL risk, in which CYP2B6 c.516G>T is related to an increased risk of AL. Aluminum 166-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 34397877-11 2021 CONCLUSION: Our meta-analysis indicated that the CYP2B6 variant is significantly associated with AL risk, in which CYP2B6 c.516G>T is related to an increased risk of AL. Aluminum 166-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-121 34116570-11 2021 Herbacetin inhibited CYP3A4, CYP2B6, CYP2C9, and CYP2E1 in a mixed manner, but non-competitively blocked CYP2D6. herbacetin 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 34181150-12 2021 Most of the cannabinoids inhibited CYP2C19, whereas CYP2D6, CYP3A4, and CYP2B6 were either not affected or only partially inhibited by the cannabinoids. Cannabinoids 139-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 34100292-6 2021 Results: CYP2B6 poor metabolizers (*6/*6) had >twofold lower methadone metabolism compared with normal/rapid metabolizers. Methadone 61-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 34100292-10 2021 Conclusion: We have described novel associations between CYP2B6 genetic variants and perioperative methadone metabolism, and associations with pain scores and PONV. Methadone 99-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 34093191-3 2021 UGT1A1*28 and CYP2B6 c.516G>T have been proposed to be related with higher toxicity by ATV and EFV, respectively. efavirenz 95-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 34093191-13 2021 Conclusion: Our findings suggest that after treatment with ATV or EFV, UGT1A1*28 and CYP2B6 c.516G>T influence the appearance of moderate-to-severe hyperbilirubinemia and CNS toxicity, respectively. Atazanavir Sulfate 59-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 85-91 34093191-0 2021 Pharmacogenetic Associations Between Atazanavir/UGT1A1*28 and Efavirenz/rs3745274 (CYP2B6) Account for Specific Adverse Reactions in Chilean Patients Undergoing Antiretroviral Therapy. Atazanavir Sulfate 37-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 35606478-5 2022 The a priori selected primary hypothesis was that patients who have CYP2B6 reduced metabolizer activity (poor or intermediate (PM/IM) vs. normal (NM) metabolizer) have lower risk of severe toxicity or cyclophosphamide treatment modification due to toxicity. Cyclophosphamide 201-217 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 34093191-0 2021 Pharmacogenetic Associations Between Atazanavir/UGT1A1*28 and Efavirenz/rs3745274 (CYP2B6) Account for Specific Adverse Reactions in Chilean Patients Undergoing Antiretroviral Therapy. efavirenz 62-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 34093191-3 2021 UGT1A1*28 and CYP2B6 c.516G>T have been proposed to be related with higher toxicity by ATV and EFV, respectively. Atazanavir Sulfate 87-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-20 35484635-5 2022 Composite CYP2B6*6/*18 genotype was significantly associated with trough plasma nevirapine levels (geometric mean (standard deviation): 4482 ng/ml (1349) of normal metabolizers vs. 4632 ng/ml (1793) of intermediate metabolizers vs. 6229 ng/ml (2549) of poor metabolizers; P < 0.001), but not the plasma HIV RNA levels, absolute lymphocyte and CD4+ T lymphocyte counts. Nevirapine 80-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 35484635-7 2022 Composite CYP2B6*6/*18 genotype correlated with plasma nevirapine levels but not immunologic and virologic responses. Nevirapine 55-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 10-16 34380920-6 2021 The mRNA expressions of the CYP2B6 and ATP-binding cassette transporter genes were decreased after 5-FU treatment. Fluorouracil 99-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 35599347-7 2022 FFA and nFFA did not induce CYP1A2; both induced CYP2B6 (up to 2.8-fold and up to 2.0-fold, respectively) and CYP3A4 (1.9- to 3.0-fold and 3.6- to 4.8-fold, respectively). Fenfluramine 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 35599347-7 2022 FFA and nFFA did not induce CYP1A2; both induced CYP2B6 (up to 2.8-fold and up to 2.0-fold, respectively) and CYP3A4 (1.9- to 3.0-fold and 3.6- to 4.8-fold, respectively). Norfenfluramine 8-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 35538637-1 2022 AIMS: Methadone metabolism and clearance are determined principally by polymorphic cytochrome P4502B6 (CYP2B6). Methadone 6-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-109 35579950-3 2022 CN06 enhances CAR-induced bioactivation of CPA (a prodrug) by provoking hepatic expression of CYP2B6, while repressing DOX-induced cytotoxicity in cardiomyocytes in vitro via stimulating Nrf2-antioxidant signaling. Cyclophosphamide 43-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 35579950-4 2022 Utilizing a multicellular co-culture model incorporating human primary hepatocytes, TNBC cells, and cardiomyocytes, we show that CN06 increased CPA/DOX-mediated TNBC cell death via CAR-dependent CYP2B6 induction and subsequent conversion of CPA to its active metabolite 4-hydroxy-CPA, while protecting against DOX-induced cardiotoxicity by selectively activating Nrf2-antioxidant signaling in cardiomyocytes but not in TNBC cells. Cyclophosphamide 144-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-201 35579950-4 2022 Utilizing a multicellular co-culture model incorporating human primary hepatocytes, TNBC cells, and cardiomyocytes, we show that CN06 increased CPA/DOX-mediated TNBC cell death via CAR-dependent CYP2B6 induction and subsequent conversion of CPA to its active metabolite 4-hydroxy-CPA, while protecting against DOX-induced cardiotoxicity by selectively activating Nrf2-antioxidant signaling in cardiomyocytes but not in TNBC cells. Doxorubicin 148-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-201 35579824-6 2022 The relative contributions of different cytochromes P450 (CYPs), mainly CYP3A4 and CYP2B6, involved in esketamine/noresketamine clearance was captured correctly in the IN-PBPK model using the DDI clinical studies of intranasal esketamine with clarithromycin and rifampicin and a published DDI study of oral esketamine with ticlopidine. noresketamine 114-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-89 35545255-6 2022 In human hepatocytes (n=3), 13cisRA, atRA and 4-oxo-13cisRA decreased OATP1B1, CYP1A2, CYP2C9, and CYP2D6 mRNA and increased CYP2B6 and CYP3A4 mRNA in a concentration-dependent manner. 13cisra 28-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 35545255-6 2022 In human hepatocytes (n=3), 13cisRA, atRA and 4-oxo-13cisRA decreased OATP1B1, CYP1A2, CYP2C9, and CYP2D6 mRNA and increased CYP2B6 and CYP3A4 mRNA in a concentration-dependent manner. Tretinoin 37-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 35545255-6 2022 In human hepatocytes (n=3), 13cisRA, atRA and 4-oxo-13cisRA decreased OATP1B1, CYP1A2, CYP2C9, and CYP2D6 mRNA and increased CYP2B6 and CYP3A4 mRNA in a concentration-dependent manner. 4-oxo-13cisra 46-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-131 35538637-15 2022 CYP2B6 polymorphisms 516G>T and 983T>C code for canonical loss of function variants, and should be assessed when considering genetic influences on clinical methadone disposition. Methadone 156-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 35538637-2 2022 Some CYP2B6 allelic variants affect methadone metabolism in vitro and disposition in vivo. Methadone 36-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-11 35538637-3 2022 We assessed methadone metabolism by CYP2B6 minor variants in vitro. Methadone 12-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-42 35538637-11 2022 CYP2B6.1 expressed with POR variants POR.28, POR.5, and P228L had lower rates of methadone metabolism than wild-type reductase. Methadone 81-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 35538637-12 2022 In vivo, methadone clinical clearance decreased linearly with the number of CYP2B6 slow metabolizer alleles, but were not different in CYP2C19 slow or rapid metabolizer phenotypes, or in carriers of the POR*28 allele. Methadone 9-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-82 35538637-13 2022 CONCLUSIONS: Several CYP2B6 and POR variants were slow metabolizers of methadone in vitro. Methadone 71-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 35538637-14 2022 Polymorphisms in CYP2B6, but not CYP2C19 or P450 reductase, affected methadone clearance in vivo. Methadone 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 35512066-0 2022 The role of CYP2B6 516G>T polymorphism on efavirenz/nevirapine toxicity. Nevirapine 52-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 35246464-6 2022 Good estimation of fm,CYP2B6 was not possible in this study due to the poor selectivity of the tested inhibitor (20 microM ticlopidine). Ticlopidine 123-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 35484370-8 2022 Ulotaront is an inducer of CYP2B6, and an inhibitor of CYP2D6, OCT1 and OCT2, while SEP-383103 is neither a CYP inducer nor a potent inhibitor of CYPs and human transporters. UNII-3K6270MG59 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 35512066-5 2022 One of the polymorphism in the CYP2B6 gene, 516G>T, particularly the 516T allele, is known to confer poor metabolism of EFV and NVP. efavirenz 120-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 35631502-1 2022 The antiplatelet agent clopidogrel is listed by the FDA as a strong clinical index inhibitor of cytochrome P450 (CYP) 2C8 and weak clinical inhibitor of CYP2B6. Clopidogrel 23-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 153-159 35293300-10 2022 Time-dependant CYP inhibition potential of ZY12201 was assessed against CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4/5 and no apparent IC50 shift was observed. zy12201 43-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-86 35399214-10 2022 On the basis of recombinant CYP studies, the carbofuran pathway was dominated by CYP3A4 (95.9%); contributions by CYP1A2 (1.3%) and CYP2B6 (2.0%) were minor. Carbofuran 45-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-138 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 2 subfamily B member 6 Homo sapiens 135-141 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 2 subfamily B member 6 Homo sapiens 135-141 35293300-9 2022 ZY12201 Ki values were 0.38, 0.25, 0.07, 0.01, 0.06, 0.02, 7.13, 0.03 and 0.003 muM for CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4/5 (substrate: testosterone) and CYP3A4/5 (substrate: midazolam), respectively. zy12201 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 96-102 35631502-0 2022 Physiologically Based Pharmacokinetic (PBPK) Modeling of Clopidogrel and Its Four Relevant Metabolites for CYP2B6, CYP2C8, CYP2C19, and CYP3A4 Drug-Drug-Gene Interaction Predictions. Clopidogrel 57-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 107-113 35115300-3 2022 The IC50,u of CBD, 7-OH CBD, THC, and 11-OH THC against CYP2B6 was 0.05, 0.34, 0.40, and 0.38 microM, respectively and against CYP2C8 was 0.28, 1.02, 0.67, and 4.37 microM, respectively. Cannabidiol 24-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 35115300-3 2022 The IC50,u of CBD, 7-OH CBD, THC, and 11-OH THC against CYP2B6 was 0.05, 0.34, 0.40, and 0.38 microM, respectively and against CYP2C8 was 0.28, 1.02, 0.67, and 4.37 microM, respectively. Dronabinol 29-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 35115300-3 2022 The IC50,u of CBD, 7-OH CBD, THC, and 11-OH THC against CYP2B6 was 0.05, 0.34, 0.40, and 0.38 microM, respectively and against CYP2C8 was 0.28, 1.02, 0.67, and 4.37 microM, respectively. Dronabinol 44-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-62 35115300-4 2022 7-COOH CBD, but not 11-COOH THC, was a weak inhibitor of CYP2B6 and 2C8. 7-cooh cbd 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 35356868-5 2022 RESULTS: When adjusted for age, gender, body mass index, and genotypes of CYP2B6 and CYP3A5, the concentration-to-dose ratio of methadone did not increase among the participants with liver fibrosis (Coefficient: 0.70; 95% CI: -2.16, 3.57; P: 0.631), even among those with advanced cirrhosis (-0.50; -4.59, 3.59; 0.810). Methadone 128-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-80 35293300-13 2022 Inhibition of CYP1A2, CYP2B6, CYP2C19, and CYP2E1 by ZY12201 was competitive, while inhibition of CYP2C8, CYP2C9, CYP2D6, and CYP3A4/5 was of mixed-mode. zy12201 53-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 35293300-14 2022 ZY12201 is a non-time-dependent inhibitor of CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4/5. zy12201 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 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 2 subfamily B member 6 Homo sapiens 6-12 35235559-0 2022 Effects of cytochrome P450 2B6 and constitutive androstane receptor genetic variation on Efavirenz plasma concentrations among HIV patients in Kenya. efavirenz 89-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-30 35235559-1 2022 The effects of genetic variation of cytochrome P450 2B6 (CYP2B6) and constitutive androstane receptor (CAR) on efavirenz (EFV) plasma concentration was evaluated among 312 HIV patients in Nairobi Kenya. efavirenz 122-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-55 34989853-7 2022 In humans, CYP3A4 and CYP3A5 followed by CYP2B6 were the main enzymes responsible for the hepatic metabolism of helenalin. helenalin 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 34989853-10 2022 It may be concluded that the metabolism of helenalin differs between rats and humans, in the latter its oxidation is catalyzed by hepatic CYP2B6, CYP3A4, CYP3A5, and CYP3A7, and extrahepatic CYP2A13. helenalin 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 35099526-0 2022 Corrigendum to: CYP2B6 Genotype and Weight Gain Differences Between Dolutegravir and Efavirenz. dolutegravir 68-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 35099526-0 2022 Corrigendum to: CYP2B6 Genotype and Weight Gain Differences Between Dolutegravir and Efavirenz. efavirenz 85-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 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 2 subfamily B member 6 Homo sapiens 6-12 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 2 subfamily B member 6 Homo sapiens 6-12 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 2 subfamily B member 6 Homo sapiens 6-12 35194103-4 2022 Strong association was observed between hepatic S-mephenytoin N-demethylase activity and CYP2B6 mRNA expression (P < 0.0001). Mephenytoin 48-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-95 35062050-13 2022 In the NAFLD model, Omeprazole maintained an expected induction of CYP1A1, however Phenytoin and Rifampicin showed elevated induction of CYP2B6 and CYP2C9 compared to healthy cultures. Phenytoin 83-92 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 35243363-6 2022 5) Age-related refinements for CPF/CPFO were primarily from ToxCast/Tox21 active hit-calls for nuclear receptors, CYP2B6 and AChE inhibition (CPFO only) associated with the metabolic pathway. O,O-diethyl O-3,5,6-trichloro-2-pyridyl phosphate 35-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 35062050-13 2022 In the NAFLD model, Omeprazole maintained an expected induction of CYP1A1, however Phenytoin and Rifampicin showed elevated induction of CYP2B6 and CYP2C9 compared to healthy cultures. Rifampin 97-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 35000550-3 2021 In vitro, CC-90001 glucuronidation was catalyzed by UGT1A9, UGT1A4, and UGT1A1, while oxidative metabolism was primarily mediated by CYP3A4/5 with minor contributions from CYP1A2, CYP2C9, CYP2B6, and CYP2D6.2. CC-90001 10-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 188-194 35387543-2 2022 Oxidation of 3 -methoxyflavone, 4 -methoxyflavone, and 3 ,4 -dimethoxyflavone and their derivatives containing 5,7-dihydroxyl groups by human cytochrome P450 (P450 or CYP) 1B1 and 2A13 was determined using LC-MS/MS systems.2. 4'-methoxyflavone 32-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-184 35387543-2 2022 Oxidation of 3 -methoxyflavone, 4 -methoxyflavone, and 3 ,4 -dimethoxyflavone and their derivatives containing 5,7-dihydroxyl groups by human cytochrome P450 (P450 or CYP) 1B1 and 2A13 was determined using LC-MS/MS systems.2. 3',4'-dimethoxyflavone 55-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-184 35387543-2 2022 Oxidation of 3 -methoxyflavone, 4 -methoxyflavone, and 3 ,4 -dimethoxyflavone and their derivatives containing 5,7-dihydroxyl groups by human cytochrome P450 (P450 or CYP) 1B1 and 2A13 was determined using LC-MS/MS systems.2. 5,7-dihydroxyl 111-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-184 35078445-12 2022 In vitro, we further confirmed that curcumin significantly downregulated the expression of AURKA, CDK1, and TOP2A genes, while significantly upregulated the expression of CYP2B6, CYP2C9, and CYP3A4 genes. Curcumin 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-177 34380995-8 2022 RESULTS: An effective methadone dose correlated with sex, BMI and the presence of ABCB1 2677GG (rs2032582) and CYP2B6 516GG (rs374527). Methadone 22-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-117 35000550-4 2021 CC-90001 in vitro inhibits CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, and CYP3A4 <= 55% at 100 muM, and the inhibition was negligible at <=30 muM. CC-90001 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 35000550-6 2021 In human hepatocytes CC-90001 is an inducer of CYP2B6 and CYP3A, with mRNA levels increased 34.4% to 52.8% relative to positive controls.4. CC-90001 21-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 35387543-2 2022 Oxidation of 3 -methoxyflavone, 4 -methoxyflavone, and 3 ,4 -dimethoxyflavone and their derivatives containing 5,7-dihydroxyl groups by human cytochrome P450 (P450 or CYP) 1B1 and 2A13 was determined using LC-MS/MS systems.2. 3-methoxyflavone 13-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-184 35115810-0 2022 The Cyp2b6 Gene Polymorphism and Phenotypic Correlation of Efavirenz-Based Combination Therapy Among the Niger Delta Ethnic Population: Implications in Modern Pharmacogenomics. efavirenz 59-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 35115810-12 2022 Genetic polymorphism of the CYP2B6 gene is prevalent among HIV/AIDs patients in the Niger Delta ethnic population on efavirenz-based HAART treatment, as the population having homozygous mutant gene or PM are >1% (6%). efavirenz 117-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. Sodium Dodecyl Sulfate 102-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-57 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. Sodium Dodecyl Sulfate 102-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-29 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. Sodium Dodecyl Sulfate 102-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-57 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. polyacrylamide 125-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-29 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. polyacrylamide 125-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-57 2603928-4 1989 The amount of aromatase P-450 (aromatase cytochrome P-450) in tissue homogenates was determined after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting by use of a polyclonal antibody directed against aromatase cytochrome P-450. polyacrylamide 125-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-57 2688634-2 1989 P450hA7 metabolized the immunosuppressant drug cyclosporin A and the dihydropyridine calcium channel antagonist nifedipine, but did not metabolize a similar dihydropyridine drug, nicardipine, nor a series of alkoxyresorufin model substrates. Cyclosporine 47-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-7 2574176-2 1989 The phenobarbital-inducible P-450 forms IIB1 and IIB2 are identical in sequence except for 14 amino acid differences within the carboxyl-terminal half of the molecule. Phenobarbital 4-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-44 2574176-6 1989 Although the published sequences of IIB1 and IIB2 are identical within the N-terminal halves of the proteins, sequence analysis of the variant cDNA revealed two amino acid substitutions in this region; Leu58----Phe and I1e114----Phe. Phenylalanine 211-214 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-40 2574176-9 1989 A IIB1 protein that possessed only the I1e114----Phe replacement catalyzed the production of all four testosterone metabolites with only slightly different product ratios compared with the parent enzyme. Phenylalanine 49-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 2-6 2574176-9 1989 A IIB1 protein that possessed only the I1e114----Phe replacement catalyzed the production of all four testosterone metabolites with only slightly different product ratios compared with the parent enzyme. Testosterone 102-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 2-6 2686430-6 1989 With respect to the effect on theophylline metabolism, quinolones inhibit specific cytochrome P-450 isozymes responsible for metabolism of methylxanthines, although there are major differences between the quinolones. Theophylline 30-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-99 2686430-6 1989 With respect to the effect on theophylline metabolism, quinolones inhibit specific cytochrome P-450 isozymes responsible for metabolism of methylxanthines, although there are major differences between the quinolones. Quinolones 55-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-99 2686430-6 1989 With respect to the effect on theophylline metabolism, quinolones inhibit specific cytochrome P-450 isozymes responsible for metabolism of methylxanthines, although there are major differences between the quinolones. methylxanthine 139-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-99 2686430-6 1989 With respect to the effect on theophylline metabolism, quinolones inhibit specific cytochrome P-450 isozymes responsible for metabolism of methylxanthines, although there are major differences between the quinolones. Quinolones 205-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-99 2813390-1 1989 The cytochrome P-450 superfamily of enzymes catalyzes the oxidative metabolism of innumerable lipophilic compounds (e.g., drugs, carcinogens, steroids). Steroids 142-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-20 2688634-2 1989 P450hA7 metabolized the immunosuppressant drug cyclosporin A and the dihydropyridine calcium channel antagonist nifedipine, but did not metabolize a similar dihydropyridine drug, nicardipine, nor a series of alkoxyresorufin model substrates. Nifedipine 112-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-7 2688634-2 1989 P450hA7 metabolized the immunosuppressant drug cyclosporin A and the dihydropyridine calcium channel antagonist nifedipine, but did not metabolize a similar dihydropyridine drug, nicardipine, nor a series of alkoxyresorufin model substrates. 1,4-dihydropyridine 69-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-7 2688634-2 1989 P450hA7 metabolized the immunosuppressant drug cyclosporin A and the dihydropyridine calcium channel antagonist nifedipine, but did not metabolize a similar dihydropyridine drug, nicardipine, nor a series of alkoxyresorufin model substrates. alkoxyresorufin 208-223 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-7 2791199-0 1989 Relative contribution of various forms of cytochrome P450 to the metabolism of benzo[a]pyrene by human liver microsomes. Benzo(a)pyrene 79-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-57 2791199-1 1989 A variety of cytochromes P450 have been implicated in the hepatic metabolism of benzo[a]pyrene (BP), including forms that are constitutively expressed and those that are highly inducible. Benzo(a)pyrene 80-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 2791199-1 1989 A variety of cytochromes P450 have been implicated in the hepatic metabolism of benzo[a]pyrene (BP), including forms that are constitutively expressed and those that are highly inducible. Benzo(a)pyrene 96-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-29 2791199-2 1989 In the present study the metabolism of BP to organic solvent-soluble derivatives by eight forms of cytochrome P450 isolated from rat liver and by a series of 11 human liver microsomal samples was investigated. Benzo(a)pyrene 39-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-114 2791199-7 1989 Of six different antibodies to forms of rat liver P450 tested, only those to P450s MC1a (P450IA2), MC1b (P450IA1) and UT1 (P450IIA1) consistently inhibited BP metabolism. Benzo(a)pyrene 156-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 50-54 2791199-9 1989 An antibody to cytochrome P450 PB3a (P450IIB1) did, however, inhibit the formation of metabolites at the 4,5- and 9,10-positions of BP by microsomal fractions of livers from one individual who had been receiving the drug phenytoin. Benzo(a)pyrene 132-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 2791199-9 1989 An antibody to cytochrome P450 PB3a (P450IIB1) did, however, inhibit the formation of metabolites at the 4,5- and 9,10-positions of BP by microsomal fractions of livers from one individual who had been receiving the drug phenytoin. Phenytoin 221-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 2791199-10 1989 These data indicate that several forms of P450 in human liver are involved in the metabolism of BP and that both constitutively expressed as well as inducible forms are important in its disposition in man. Benzo(a)pyrene 96-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-46 2496984-2 1989 The difference in pentoxyresorufin O-dealkylating activity observed in a reconstituted system containing dilauroylglycerophosphocholine (Lau2GroPCho) or distearoylglycerophosphocholine (Ste2GroPCho) was used as a model to study the role of phospholipids in the reconstituted cytochrome P-450b (IIB1) system. dilauroylglycerophosphocholine 105-135 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 294-298 2815828-7 1989 RP 52028 appears to be an inducer of cytochrome P-450 II B1 (P-450b) and related enzymic activities; i.e. benzphetamine, ethylmorphine and aminopyrine demethylation. Benzphetamine 106-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-59 2815828-7 1989 RP 52028 appears to be an inducer of cytochrome P-450 II B1 (P-450b) and related enzymic activities; i.e. benzphetamine, ethylmorphine and aminopyrine demethylation. Ethylmorphine 121-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-59 2815828-7 1989 RP 52028 appears to be an inducer of cytochrome P-450 II B1 (P-450b) and related enzymic activities; i.e. benzphetamine, ethylmorphine and aminopyrine demethylation. Aminopyrine 139-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-59 2496984-2 1989 The difference in pentoxyresorufin O-dealkylating activity observed in a reconstituted system containing dilauroylglycerophosphocholine (Lau2GroPCho) or distearoylglycerophosphocholine (Ste2GroPCho) was used as a model to study the role of phospholipids in the reconstituted cytochrome P-450b (IIB1) system. 1,2-distearoyllecithin 153-184 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 294-298 2667528-4 1989 This technique also resulted in the discovery of a new pathway of ethanol metabolism in the microsomes involving an ethanol-specific cytochrome P-450 (P450IIE1), which has now been confirmed in man. Ethanol 66-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-149 2540088-1 1989 Aromatase, 17 alpha-hydroxylase, and cholesterol side-chain cleavage P-450 cytochromes (P-450AROM, P-450(17 alpha,) and P-450SCC, respectively) were immunohistochemically localized in nine granulosa cell tumors, 15 thecomas, ten Sertoli-Leydig cell tumors, two steroid cell tumors, five fibromas, and five sclerosing stromal tumors. Cholesterol 37-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-74 2540088-3 1989 In the steroid cell tumors, all the P-450 cytochromes were intensely stained. Steroids 7-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-41 2540752-1 1989 The phosphorylation of the two major phenobarbital-inducible cytochrome P450 isoenzymes IIB1 and IIB2 was increased in hepatocytes by the action of the membrane permeating cAMP derivatives N6-dibutyryl-cAMP and 8-thiomethyl-cAMP. Phenobarbital 37-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 2540752-1 1989 The phosphorylation of the two major phenobarbital-inducible cytochrome P450 isoenzymes IIB1 and IIB2 was increased in hepatocytes by the action of the membrane permeating cAMP derivatives N6-dibutyryl-cAMP and 8-thiomethyl-cAMP. Cyclic AMP 172-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 2540752-1 1989 The phosphorylation of the two major phenobarbital-inducible cytochrome P450 isoenzymes IIB1 and IIB2 was increased in hepatocytes by the action of the membrane permeating cAMP derivatives N6-dibutyryl-cAMP and 8-thiomethyl-cAMP. n6-dibutyryl-camp 189-206 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 2540752-1 1989 The phosphorylation of the two major phenobarbital-inducible cytochrome P450 isoenzymes IIB1 and IIB2 was increased in hepatocytes by the action of the membrane permeating cAMP derivatives N6-dibutyryl-cAMP and 8-thiomethyl-cAMP. 8-(methylthio)cyclic 3',5'-adenosine monophosphate 211-228 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92 2540752-2 1989 Under these conditions the dealkylation of 7-pentoxyresorufin, a selective substrate of cytochrome P450IIB1 and P450IIB2 was markedly reduced. pentoxyresorufin 43-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-107 2667528-4 1989 This technique also resulted in the discovery of a new pathway of ethanol metabolism in the microsomes involving an ethanol-specific cytochrome P-450 (P450IIE1), which has now been confirmed in man. Ethanol 66-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-159 2667528-4 1989 This technique also resulted in the discovery of a new pathway of ethanol metabolism in the microsomes involving an ethanol-specific cytochrome P-450 (P450IIE1), which has now been confirmed in man. Ethanol 116-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 144-149 2667528-4 1989 This technique also resulted in the discovery of a new pathway of ethanol metabolism in the microsomes involving an ethanol-specific cytochrome P-450 (P450IIE1), which has now been confirmed in man. Ethanol 116-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-159 2667528-5 1989 P450IIE1 contributes not only to the metabolic tolerance to ethanol, but also explains the enhanced susceptibility of the alcoholic to many ubiquitous xenobiotic agents. Ethanol 60-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-8 2535970-0 1989 Phosphorylation of carcinogen metabolizing enzymes: regulation of the phosphorylation status of the major phenobarbital inducible cytochromes P-450 in hepatocytes. Phenobarbital 106-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 142-147 2535970-1 1989 We present data showing that the major phenobarbital inducible cytochromes P-450 (cytochrome P-450IIB1 and cytochrome P-450IIB2) were phosphorylated in intact hepatocytes. Phenobarbital 39-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-80 2598298-6 1989 Carbon monoxide (100%) caused an inhibition of about 37% and 44% for P-450 and b5, respectively. Carbon Monoxide 0-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-81 2535970-1 1989 We present data showing that the major phenobarbital inducible cytochromes P-450 (cytochrome P-450IIB1 and cytochrome P-450IIB2) were phosphorylated in intact hepatocytes. Phenobarbital 39-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-102 2535970-3 1989 Most importantly the phosphorylation status of cytochromes P-450 was shown to change in the hepatocytes after treatment with glucagon, which is known to increase the level of cAMP in hepatocytes. Glucagon 125-133 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-64 2535970-3 1989 Most importantly the phosphorylation status of cytochromes P-450 was shown to change in the hepatocytes after treatment with glucagon, which is known to increase the level of cAMP in hepatocytes. Cyclic AMP 175-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-64 3202862-1 1988 2,3,7,8-Tetrachlorodibenzo-p-dioxin exhibits antiestrogenic activity and induces cytochromes P-450 in estrogen-dependent MCF-7 human breast-cancer cells. Polychlorinated Dibenzodioxins 0-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-98 3144269-6 1988 The decrease of reductase supported P-450 reduction velocity in microsomes at high salt concentration (I greater than 222 mM) indicates the dominant electrostatic character of P-450/reductase interaction. Salts 83-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 176-191 3243766-3 1988 From the results of SDS-PAGE, the molecular weights of P-450-HM1, P-450-HM2, and P-450-HM3 were estimated to be 51,000, 54,000, and 52,000, respectively. Sodium Dodecyl Sulfate 20-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-64 3243766-5 1988 In a reconstituted system, P-450-HM1 showed the highest catalytic activities of nifedipine and (S)- or (R)-nilvadipine oxidases. Nifedipine 80-90 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-36 3243766-5 1988 In a reconstituted system, P-450-HM1 showed the highest catalytic activities of nifedipine and (S)- or (R)-nilvadipine oxidases. (s)- or (r)-nilvadipine 95-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-36 3183973-1 1988 Chronic ethanol consumption results in the induction of a specific hepatic cytochrome P-450 (P450IIE1). Ethanol 8-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-91 3183973-1 1988 Chronic ethanol consumption results in the induction of a specific hepatic cytochrome P-450 (P450IIE1). Ethanol 8-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-101 3183973-2 1988 However, since compounds other than ethanol (i.e., acetone) can also serve as P450IIE1 inducers, and since ethanol given with a normal fat-containing (35% of energy) diet is associated with acetonemia, hepatic steatosis and decreased body weight gain, the question has been raised whether induction is mediated specifically by ethanol or whether it might represent a nonspecific response to these other factors. Acetone 51-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 78-86 3183973-4 1988 By administering ethanol in the drinking water, or as part of a low-fat (5% of energy) liquid diet, a significant induction of P450IIE1 and of the activities of the microsomal ethanol oxidizing system and p-nitrophenol hydroxylase was demonstrated in the absence of any significant increase in blood acetone with minimal increase in liver total lipids. Ethanol 17-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-135 3183973-4 1988 By administering ethanol in the drinking water, or as part of a low-fat (5% of energy) liquid diet, a significant induction of P450IIE1 and of the activities of the microsomal ethanol oxidizing system and p-nitrophenol hydroxylase was demonstrated in the absence of any significant increase in blood acetone with minimal increase in liver total lipids. Water 41-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-135 3183973-5 1988 Induction of P450IIE1 was comparable with the low or normal fat-containing diets, but MEOS activity rose more with the latter, possibly reflecting a potentiating effect of dietary fat on ethanol oxidation by P-450 enzymes other than P450IIE1. Ethanol 187-194 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 208-213 3183973-8 1988 Thus, the induction of P450IIE1 after chronic ethanol consumption can be attributed to ethanol itself, but dietary fat can potentiate the induction of the microsomal ethanol oxidizing system and of p-nitrophenol hydroxylase. Ethanol 46-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-31 3183973-8 1988 Thus, the induction of P450IIE1 after chronic ethanol consumption can be attributed to ethanol itself, but dietary fat can potentiate the induction of the microsomal ethanol oxidizing system and of p-nitrophenol hydroxylase. Ethanol 87-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-31 3183973-8 1988 Thus, the induction of P450IIE1 after chronic ethanol consumption can be attributed to ethanol itself, but dietary fat can potentiate the induction of the microsomal ethanol oxidizing system and of p-nitrophenol hydroxylase. Ethanol 87-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-31 3185539-1 1988 Homologous 1-naphtoxyalcanthiols of the type 1-C10H7O(CH2)nSH (n = 2-7) are used for structural studies of the microsomal cytochrome P450 active centre. 1-naphtoxyalcanthiols 11-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-137 3185539-2 1988 It was found that the strongest complex of thiol with P450 is formed for n = 3. Sulfhydryl Compounds 43-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 3185539-3 1988 Microsomal oxidation of P450 substrates aminopyrine and benz(a) pyrene is inhibited by the 1-naphtoxyalcanthiols studied. Aminopyrine 40-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 3185539-3 1988 Microsomal oxidation of P450 substrates aminopyrine and benz(a) pyrene is inhibited by the 1-naphtoxyalcanthiols studied. Benzo(a)pyrene 56-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 3185539-3 1988 Microsomal oxidation of P450 substrates aminopyrine and benz(a) pyrene is inhibited by the 1-naphtoxyalcanthiols studied. 1-naphtoxyalcanthiols 91-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-28 3185539-5 1988 The interaction of this thiol (n = 3) with both the heme group of P450 and the hydrophobic substrate zone is supposed and the distance between these points was estimated. Sulfhydryl Compounds 24-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 3185539-5 1988 The interaction of this thiol (n = 3) with both the heme group of P450 and the hydrophobic substrate zone is supposed and the distance between these points was estimated. Heme 52-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 66-70 3050970-2 1988 The antibodies used in the immunohistochemical analyses were monoclonal antibodies (MAbs) 1-7-1 and 2-66-3, prepared against the 3-methylcholanthrene-induced and phenobarbital-induced rat liver P-450, respectively. Phenobarbital 162-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 194-199 3050970-7 1988 These results demonstrate that maternal cigarette smoking induces in the trophoblastic layer of the placenta a cytochrome P-450 form which is detectable immunohistochemically with the monoclonal antibody to 3-methylcholanthrene-inducible P-450 in rat liver. Methylcholanthrene 207-227 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-127 3378062-0 1988 [Isolation and characteristics of coumarin-specific cytochrome P-450 (P-450Cho) from liver microsomes of DBA/2N mice, induced with pyrazole]. coumarin 34-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-68 3378062-0 1988 [Isolation and characteristics of coumarin-specific cytochrome P-450 (P-450Cho) from liver microsomes of DBA/2N mice, induced with pyrazole]. pyrazole 131-139 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-68 3378062-1 1988 Coumarin-specific cytochrome P-450 (P-450Coh) has been isolated from liver microsomes of DBA/2N mice induced with pyrazole. pyrazole 114-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-34 3378062-1 1988 Coumarin-specific cytochrome P-450 (P-450Coh) has been isolated from liver microsomes of DBA/2N mice induced with pyrazole. pyrazole 114-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-44 3378062-3 1988 At the final step of P-450Coh purification, variously substituted Sepharoses (hydroxyphenyl-, cholate-, aminooctyl- and t-cytochrome-b5-) were used. Sepharose 66-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-29 3378062-5 1988 According to SDS gel electrophoresis data, the purity of P-450Coh was 95% and Mr was 50,000 Da. Sodium Dodecyl Sulfate 13-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-65 3378062-7 1988 At saturating concentrations of coumarin, more than 90% of P-450Coh are represented by the high spin form. coumarin 32-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-67 3502608-3 1987 The levels of mRNAs for the rate-limiting enzymes in the conversion of cholesterol into progesterone, namely cholesterol side-chain cleavage cytochrome P-450 and its electron donor, adrenodoxin, were higher in corpora lutea than in follicles. Cholesterol 71-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-157 2856164-8 1988 The RA metabolism in rat epidermal microsomes shows the typical characteristics of a cytochrome-P-450 (P450)-dependent enzyme system, i.e. a requirement for NADPH and oxygen and inhibition by CO and SKF-525A. Tretinoin 4-6 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-107 2856164-8 1988 The RA metabolism in rat epidermal microsomes shows the typical characteristics of a cytochrome-P-450 (P450)-dependent enzyme system, i.e. a requirement for NADPH and oxygen and inhibition by CO and SKF-525A. NADP 157-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-107 2856164-8 1988 The RA metabolism in rat epidermal microsomes shows the typical characteristics of a cytochrome-P-450 (P450)-dependent enzyme system, i.e. a requirement for NADPH and oxygen and inhibition by CO and SKF-525A. Oxygen 167-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-107 2856164-8 1988 The RA metabolism in rat epidermal microsomes shows the typical characteristics of a cytochrome-P-450 (P450)-dependent enzyme system, i.e. a requirement for NADPH and oxygen and inhibition by CO and SKF-525A. Proadifen 199-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-107 2856164-9 1988 Ketoconazole and miconazole, imidazole antifungal agents and inhibitors of some fungal and mammalian P450-dependent enzymes, inhibit in vitro RA metabolism by rat epidermal microsomes. Miconazole 17-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 2856164-9 1988 Ketoconazole and miconazole, imidazole antifungal agents and inhibitors of some fungal and mammalian P450-dependent enzymes, inhibit in vitro RA metabolism by rat epidermal microsomes. Tretinoin 142-144 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-105 2856164-13 1988 Our data show a P450-dependent RA metabolism in rat epidermal microsomes and suggest that ketoconazole may prove to be effective in maintaining biologically active levels of RA in epidermal cells. Tretinoin 31-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 3654968-2 1987 In rat enterocytes, we found two dexamethasone-inducible proteins related to the steroid-inducible liver cytochromes P-450. Dexamethasone 33-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-122 3654968-2 1987 In rat enterocytes, we found two dexamethasone-inducible proteins related to the steroid-inducible liver cytochromes P-450. Steroids 81-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 117-122 3663605-2 1987 Water proton relaxation rates of various complexes of cholesterol side chain cleavage cytochrome P-450 (-450scc) were investigated to gain information about the structure and dynamics of the steroid binding site. Water 0-5 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-111 3663605-2 1987 Water proton relaxation rates of various complexes of cholesterol side chain cleavage cytochrome P-450 (-450scc) were investigated to gain information about the structure and dynamics of the steroid binding site. Cholesterol 54-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-111 3663605-2 1987 Water proton relaxation rates of various complexes of cholesterol side chain cleavage cytochrome P-450 (-450scc) were investigated to gain information about the structure and dynamics of the steroid binding site. Steroids 191-198 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-111 3142109-1 1987 Microsomal estrogen synthetase (aromatase) cytochrome P-450 was purified from fresh human placental microsomes by monoclonal anti-aromatase P-450 antibody-Sepharose 4B chromatography. Sepharose 155-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-59 3142109-1 1987 Microsomal estrogen synthetase (aromatase) cytochrome P-450 was purified from fresh human placental microsomes by monoclonal anti-aromatase P-450 antibody-Sepharose 4B chromatography. Sepharose 155-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-145 3142109-2 1987 The purified P-450 showed a single band of 55 kDa on SDS-polyacrylamide gel electrophoresis and the aromatase specific activity on reconstitution was 70 nmol/min/mg protein. Sodium Dodecyl Sulfate 53-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-18 3142109-2 1987 The purified P-450 showed a single band of 55 kDa on SDS-polyacrylamide gel electrophoresis and the aromatase specific activity on reconstitution was 70 nmol/min/mg protein. polyacrylamide 57-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-18 3142109-3 1987 The purified P-450 was stable with a t 1/2 of approximately 2 years on storage at -90 degrees C and showed Km = 43 nM for androstenedione aromatization. Androstenedione 122-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-18 3142109-4 1987 However, it was unstable under spectral measurement conditions in the presence of sodium dithionite and carbon monoxide and the carbon monoxide difference spectra showed a maximum at 450 nm and a specific content of 9.1 nmol of P-450/mg protein, giving a turnover number of approximately 7.7 per min for the purified aromatase. Dithionite 82-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 228-233 3142109-4 1987 However, it was unstable under spectral measurement conditions in the presence of sodium dithionite and carbon monoxide and the carbon monoxide difference spectra showed a maximum at 450 nm and a specific content of 9.1 nmol of P-450/mg protein, giving a turnover number of approximately 7.7 per min for the purified aromatase. Carbon Monoxide 104-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 228-233 3142109-4 1987 However, it was unstable under spectral measurement conditions in the presence of sodium dithionite and carbon monoxide and the carbon monoxide difference spectra showed a maximum at 450 nm and a specific content of 9.1 nmol of P-450/mg protein, giving a turnover number of approximately 7.7 per min for the purified aromatase. Carbon Monoxide 128-143 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 228-233 2882985-0 1987 Biotransformation of caffeine, paraxanthine, theophylline, and theobromine by polycyclic aromatic hydrocarbon-inducible cytochrome(s) P-450 in human liver microsomes. Caffeine 21-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 2882985-0 1987 Biotransformation of caffeine, paraxanthine, theophylline, and theobromine by polycyclic aromatic hydrocarbon-inducible cytochrome(s) P-450 in human liver microsomes. 1,7-dimethylxanthine 31-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 2882985-0 1987 Biotransformation of caffeine, paraxanthine, theophylline, and theobromine by polycyclic aromatic hydrocarbon-inducible cytochrome(s) P-450 in human liver microsomes. Theophylline 45-57 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 2882985-0 1987 Biotransformation of caffeine, paraxanthine, theophylline, and theobromine by polycyclic aromatic hydrocarbon-inducible cytochrome(s) P-450 in human liver microsomes. Theobromine 63-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 2882985-0 1987 Biotransformation of caffeine, paraxanthine, theophylline, and theobromine by polycyclic aromatic hydrocarbon-inducible cytochrome(s) P-450 in human liver microsomes. Polycyclic Aromatic Hydrocarbons 78-109 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 2856164-13 1988 Our data show a P450-dependent RA metabolism in rat epidermal microsomes and suggest that ketoconazole may prove to be effective in maintaining biologically active levels of RA in epidermal cells. Ketoconazole 90-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 2856164-13 1988 Our data show a P450-dependent RA metabolism in rat epidermal microsomes and suggest that ketoconazole may prove to be effective in maintaining biologically active levels of RA in epidermal cells. Tretinoin 174-176 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-20 3500726-0 1987 Interactions of imidazole antifungal agents with purified cytochrome P-450 proteins. imidazole 16-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-74 3500726-1 1987 The imidazole N-substituted antifungal agents ketoconazole, miconazole and clotrimazole have been shown to be potent inhibitors of oxidative metabolism by both a phenobarbital-induced cytochrome P-450 (P-450b) and a 3-methylcholanthrene-induced cytochrome P-448-protein (P-450c) in reconstituted systems. imidazole n 4-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-200 3500726-1 1987 The imidazole N-substituted antifungal agents ketoconazole, miconazole and clotrimazole have been shown to be potent inhibitors of oxidative metabolism by both a phenobarbital-induced cytochrome P-450 (P-450b) and a 3-methylcholanthrene-induced cytochrome P-448-protein (P-450c) in reconstituted systems. Ketoconazole 46-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-200 3500726-1 1987 The imidazole N-substituted antifungal agents ketoconazole, miconazole and clotrimazole have been shown to be potent inhibitors of oxidative metabolism by both a phenobarbital-induced cytochrome P-450 (P-450b) and a 3-methylcholanthrene-induced cytochrome P-448-protein (P-450c) in reconstituted systems. Miconazole 60-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-200 3500726-1 1987 The imidazole N-substituted antifungal agents ketoconazole, miconazole and clotrimazole have been shown to be potent inhibitors of oxidative metabolism by both a phenobarbital-induced cytochrome P-450 (P-450b) and a 3-methylcholanthrene-induced cytochrome P-448-protein (P-450c) in reconstituted systems. Clotrimazole 75-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-200 3500726-1 1987 The imidazole N-substituted antifungal agents ketoconazole, miconazole and clotrimazole have been shown to be potent inhibitors of oxidative metabolism by both a phenobarbital-induced cytochrome P-450 (P-450b) and a 3-methylcholanthrene-induced cytochrome P-448-protein (P-450c) in reconstituted systems. Methylcholanthrene 216-236 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 195-200 2963808-1 1987 P-450 human-2 is a human cytochrome P-450 that is immunochemically related to a constitutive male-specific cytochrome P-450 (P-450-male) and the phenobarbital-inducible P-450b/e in rat liver. Phenobarbital 145-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 2963808-1 1987 P-450 human-2 is a human cytochrome P-450 that is immunochemically related to a constitutive male-specific cytochrome P-450 (P-450-male) and the phenobarbital-inducible P-450b/e in rat liver. Phenobarbital 145-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-41 2963808-1 1987 P-450 human-2 is a human cytochrome P-450 that is immunochemically related to a constitutive male-specific cytochrome P-450 (P-450-male) and the phenobarbital-inducible P-450b/e in rat liver. Phenobarbital 145-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 36-41 2963808-4 1987 The N-terminal 11-amino-acid sequence was in agreement with the protein sequence of P-450 human-2. 11-amino-acid 15-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-89 3445541-1 1987 The properties of tert-butanol, sodium deoxycholate, guanidine and mersalyl to convert microsomal cytochromes P-450 and P-448 into enzymatically inactive form P-420 were studied under comparable conditions. tert-Butyl Alcohol 18-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-125 3445541-1 1987 The properties of tert-butanol, sodium deoxycholate, guanidine and mersalyl to convert microsomal cytochromes P-450 and P-448 into enzymatically inactive form P-420 were studied under comparable conditions. Deoxycholic Acid 32-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-125 3445541-1 1987 The properties of tert-butanol, sodium deoxycholate, guanidine and mersalyl to convert microsomal cytochromes P-450 and P-448 into enzymatically inactive form P-420 were studied under comparable conditions. Guanidine 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-125 3445541-1 1987 The properties of tert-butanol, sodium deoxycholate, guanidine and mersalyl to convert microsomal cytochromes P-450 and P-448 into enzymatically inactive form P-420 were studied under comparable conditions. Mersalyl 67-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-125 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 2 subfamily B member 6 Homo sapiens 11-20 3675576-3 1987 In a reconstituted system, P-450-ALC oxidizes ethanol and aniline at turnover rates (12.2 and 7.3 nmol min-1, respectively) 10-fold greater than two other human P-450 isozymes (termed P-450-B and P-450-C) purified from the same liver. Ethanol 46-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-36 3675576-3 1987 In a reconstituted system, P-450-ALC oxidizes ethanol and aniline at turnover rates (12.2 and 7.3 nmol min-1, respectively) 10-fold greater than two other human P-450 isozymes (termed P-450-B and P-450-C) purified from the same liver. aniline 58-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-36 3675576-4 1987 Both P-450-ALC and P-450-C effectively demethylate N-nitrosodimethylamine (NDMA) at low substrate concentrations (0.5 mM), especially in the presence of cytochrome b5. demethylate n-nitrosodimethylamine 39-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-14 3675576-4 1987 Both P-450-ALC and P-450-C effectively demethylate N-nitrosodimethylamine (NDMA) at low substrate concentrations (0.5 mM), especially in the presence of cytochrome b5. Dimethylnitrosamine 75-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-14 3597396-2 1987 19-Norandrostenedione and androstenedione are shown to be metabolized by purified, reconstituted human placental aromatase cytochrome P-450. 19-norandrostenedione 0-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 3597396-2 1987 19-Norandrostenedione and androstenedione are shown to be metabolized by purified, reconstituted human placental aromatase cytochrome P-450. Androstenedione 6-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 134-139 3107803-7 1987 Nine of the MAbs were tested for cross-reactivity with rat liver clofibrate-induced P-450, rat liver pregnenolone-16-alpha-carbonitrile-induced P-450, and a human liver P-450. Clofibrate 65-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-89 3107803-13 1987 In reconstituted systems containing phospholipid and NADPH-cytochrome P-450 reductase, MAb 1-91-3 inhibited aniline p-hydroxylase activity of purified ethanol-induced P-450et and acetone-induced P-450 by more than 90%. Ethanol 151-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-75 3107803-14 1987 Nitrosodimethylamine demethylase activity of acetone-induced rat microsomes was inhibited by the various MAbs up to 77% and the activity of the purified acetone-induced P-450 was inhibited up to 92%. Acetone 45-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-174 3107803-14 1987 Nitrosodimethylamine demethylase activity of acetone-induced rat microsomes was inhibited by the various MAbs up to 77% and the activity of the purified acetone-induced P-450 was inhibited up to 92%. Acetone 153-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-174 2882985-8 1987 Furthermore, in low activity livers, more than 80% of paraxanthine 8-hydroxylation was mediated by an isozyme of cytochrome P-450 insensitive to inhibition by alpha-naphthoflavone. 1,7-dimethylxanthine 54-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-129 2882985-10 1987 Taken together, the data provide a rationale for a potential in vivo marker of polycyclic aromatic hydrocarbon-inducible cytochrome P-450 activity based on a urinary metabolite ratio of paraxanthine 7-demethylation to 8-hydroxylation products after caffeine intake. 1,7-dimethylxanthine 186-198 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-137 2882985-10 1987 Taken together, the data provide a rationale for a potential in vivo marker of polycyclic aromatic hydrocarbon-inducible cytochrome P-450 activity based on a urinary metabolite ratio of paraxanthine 7-demethylation to 8-hydroxylation products after caffeine intake. Caffeine 249-257 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-137 3502608-3 1987 The levels of mRNAs for the rate-limiting enzymes in the conversion of cholesterol into progesterone, namely cholesterol side-chain cleavage cytochrome P-450 and its electron donor, adrenodoxin, were higher in corpora lutea than in follicles. Progesterone 88-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-157 3502608-3 1987 The levels of mRNAs for the rate-limiting enzymes in the conversion of cholesterol into progesterone, namely cholesterol side-chain cleavage cytochrome P-450 and its electron donor, adrenodoxin, were higher in corpora lutea than in follicles. Cholesterol 109-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-157 3502608-4 1987 Conversely the levels of mRNA specific for the key regulatory enzyme in the conversion of pregnenolone or progesterone to androgen, namely 17 alpha-hydroxylase cytochrome P-450, were high in all antral follicles examined but were low in young corpora lutea and undetectable in more mature corpora lutea. Pregnenolone 90-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-176 3502608-4 1987 Conversely the levels of mRNA specific for the key regulatory enzyme in the conversion of pregnenolone or progesterone to androgen, namely 17 alpha-hydroxylase cytochrome P-450, were high in all antral follicles examined but were low in young corpora lutea and undetectable in more mature corpora lutea. Progesterone 106-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 171-176 3690723-1 1987 Distinct and different molecular structural features are manifested by substrates, inhibitors and inducers of the two families of liver microsomal enzymes, the phenobarbital-induced cytochromes P-450 and the 3-methylcholanthrene-induced cytochromes P-448. Phenobarbital 160-173 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 194-199 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. Alcohols 114-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-22 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. Alcohols 114-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-98 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. Ethanol 182-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-22 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. Ethanol 182-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-98 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. NADP 194-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-22 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. NADP 194-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-98 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. h+----acetaldehyde 200-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-22 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. h+----acetaldehyde 200-218 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-98 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. 2h2o 220-224 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-22 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. 2h2o 220-224 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-98 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. NADP 225-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-22 3304205-1 1987 Of the family of P-450 cytochromes occurring in rabbit liver microsomes, only isozyme 3a (P-450ALC) is induced by alcohol administration and is effective in catalyzing the reaction: ethanol +02+NADPH+H+----acetaldehyde +2H2O+NADP+. NADP 225-230 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-98 3304205-2 1987 As judged by immunochemical quantitation, P-450ALC is also induced in the animals by other diverse agents, including imidazole, trichloroethylene, acetone, pyrazole, and isoniazid. imidazole 117-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-50 3304205-2 1987 As judged by immunochemical quantitation, P-450ALC is also induced in the animals by other diverse agents, including imidazole, trichloroethylene, acetone, pyrazole, and isoniazid. Trichloroethylene 128-145 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-50 3304205-2 1987 As judged by immunochemical quantitation, P-450ALC is also induced in the animals by other diverse agents, including imidazole, trichloroethylene, acetone, pyrazole, and isoniazid. Acetone 147-154 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-50 3304205-2 1987 As judged by immunochemical quantitation, P-450ALC is also induced in the animals by other diverse agents, including imidazole, trichloroethylene, acetone, pyrazole, and isoniazid. pyrazole 156-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-50 3304205-2 1987 As judged by immunochemical quantitation, P-450ALC is also induced in the animals by other diverse agents, including imidazole, trichloroethylene, acetone, pyrazole, and isoniazid. Isoniazid 170-179 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-50 3304205-4 1987 P-450ALC catalyzes the activation of foreign compounds such as acetaminophen, various nitrosamines, and carbon tetrachloride and is therefore believed to play an important role in the enhanced toxicity of these substances accompanying alcohol administration. Acetaminophen 63-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-8 3304205-4 1987 P-450ALC catalyzes the activation of foreign compounds such as acetaminophen, various nitrosamines, and carbon tetrachloride and is therefore believed to play an important role in the enhanced toxicity of these substances accompanying alcohol administration. Nitrosamines 86-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-8 3304205-4 1987 P-450ALC catalyzes the activation of foreign compounds such as acetaminophen, various nitrosamines, and carbon tetrachloride and is therefore believed to play an important role in the enhanced toxicity of these substances accompanying alcohol administration. Carbon Tetrachloride 104-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-8 3304205-4 1987 P-450ALC catalyzes the activation of foreign compounds such as acetaminophen, various nitrosamines, and carbon tetrachloride and is therefore believed to play an important role in the enhanced toxicity of these substances accompanying alcohol administration. Alcohols 235-242 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-8 3690723-3 1987 In contrast, substrates of the cytochromes P-450 have greater conformational freedom and an ability to bind at more than one point of attachment, as a result of possession of certain characteristic functions, namely, a carbonyl and/or amine moiety coupled with an iso-propyl group, or similar function of equivalent shape and hydrophobicity. Amines 235-240 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-48 3690723-4 1987 The implications are that the binding sites of cytochromes P-448 contain a number of hydrophobic aromatic amino acid residues orientated so as to allow occupation by similar substrates containing co-planar aromatic rings, whereas those of the phenobarbital-induced cytochromes P-450 contain hydrophilic amino acid residues capable of hydrogen bonding to greater than C = O moieties and at least one leucine or valine residue, as these contain the complementary isopropyl function. Amino Acids, Aromatic 97-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 277-282 3690723-4 1987 The implications are that the binding sites of cytochromes P-448 contain a number of hydrophobic aromatic amino acid residues orientated so as to allow occupation by similar substrates containing co-planar aromatic rings, whereas those of the phenobarbital-induced cytochromes P-450 contain hydrophilic amino acid residues capable of hydrogen bonding to greater than C = O moieties and at least one leucine or valine residue, as these contain the complementary isopropyl function. Phenobarbital 243-256 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 277-282 3690723-4 1987 The implications are that the binding sites of cytochromes P-448 contain a number of hydrophobic aromatic amino acid residues orientated so as to allow occupation by similar substrates containing co-planar aromatic rings, whereas those of the phenobarbital-induced cytochromes P-450 contain hydrophilic amino acid residues capable of hydrogen bonding to greater than C = O moieties and at least one leucine or valine residue, as these contain the complementary isopropyl function. Hydrogen 334-342 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 277-282 3690723-4 1987 The implications are that the binding sites of cytochromes P-448 contain a number of hydrophobic aromatic amino acid residues orientated so as to allow occupation by similar substrates containing co-planar aromatic rings, whereas those of the phenobarbital-induced cytochromes P-450 contain hydrophilic amino acid residues capable of hydrogen bonding to greater than C = O moieties and at least one leucine or valine residue, as these contain the complementary isopropyl function. Leucine 399-406 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 277-282 3690723-4 1987 The implications are that the binding sites of cytochromes P-448 contain a number of hydrophobic aromatic amino acid residues orientated so as to allow occupation by similar substrates containing co-planar aromatic rings, whereas those of the phenobarbital-induced cytochromes P-450 contain hydrophilic amino acid residues capable of hydrogen bonding to greater than C = O moieties and at least one leucine or valine residue, as these contain the complementary isopropyl function. Valine 410-416 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 277-282 3320558-3 1987 Partially purified aromatase in each form was immunoaffinity chromatographed to give a single band (SDS-PAGE) cytochrome P-450 of 55 kDa, utilizing a mouse monoclonal anti-human placental aromatase cytochrome P-450 IgGi (MAb3-2C2) which is capable of suppressing placental aromatase activity. Sodium Dodecyl Sulfate 100-103 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-126 3681487-1 1987 P(1)450 and P(3)450 are the two members of the dioxin-inducible P450 gene family, one of at least eight families in the entire P450 gene superfamily. Dioxins 47-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 64-68 3681487-1 1987 P(1)450 and P(3)450 are the two members of the dioxin-inducible P450 gene family, one of at least eight families in the entire P450 gene superfamily. Dioxins 47-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 127-131 3320558-4 1987 The purified cytochrome P-450 showed specific aromatase activity of 25-30 nmol/min per mg with Km of 20-30 nM for androstenedione on reconstitution with NADPH-cyt P-450 reductase and dilauroyl L-alpha-phosphatidylcholine. Androstenedione 114-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-29 3320558-4 1987 The purified cytochrome P-450 showed specific aromatase activity of 25-30 nmol/min per mg with Km of 20-30 nM for androstenedione on reconstitution with NADPH-cyt P-450 reductase and dilauroyl L-alpha-phosphatidylcholine. Androstenedione 114-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 163-168 3320558-4 1987 The purified cytochrome P-450 showed specific aromatase activity of 25-30 nmol/min per mg with Km of 20-30 nM for androstenedione on reconstitution with NADPH-cyt P-450 reductase and dilauroyl L-alpha-phosphatidylcholine. dilauroyl l-alpha-phosphatidylcholine 183-220 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-29 3795953-2 1986 Steroid products and their analogs induce oxygen-mediated damage of microsomal P-450 activities of cultured Leydig cells, whereas testosterone acetate protects P-450 from this damage [1]. Steroids 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-84 3795953-10 1986 Therefore, the protection of P-450 in cultured Leydig cells probably results from binding of testosterone acetate at the active site of P-450. testosterone acetate 93-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-34 3795953-2 1986 Steroid products and their analogs induce oxygen-mediated damage of microsomal P-450 activities of cultured Leydig cells, whereas testosterone acetate protects P-450 from this damage [1]. Oxygen 42-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-84 3795953-10 1986 Therefore, the protection of P-450 in cultured Leydig cells probably results from binding of testosterone acetate at the active site of P-450. testosterone acetate 93-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 136-141 3795953-2 1986 Steroid products and their analogs induce oxygen-mediated damage of microsomal P-450 activities of cultured Leydig cells, whereas testosterone acetate protects P-450 from this damage [1]. testosterone acetate 130-150 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 160-165 3795953-3 1986 Progesterone and 17 alpha-hydroxyprogesterone, the enzyme"s substrates, bound stoichiometrically to microsomal P-450 with unusually high affinity (Kd = 23-51 nM). Progesterone 0-12 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-116 3795953-3 1986 Progesterone and 17 alpha-hydroxyprogesterone, the enzyme"s substrates, bound stoichiometrically to microsomal P-450 with unusually high affinity (Kd = 23-51 nM). 17-alpha-Hydroxyprogesterone 17-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 111-116 3795953-6 1986 Several steroids which have varying effects on damage of P-450 in cultured Leydig cells inhibited substrate binding similarly (KdI = 3.8-12.8 microM). Steroids 8-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-62 3795953-8 1986 The results suggest that steroid binding to P-450 is necessary but not sufficient to increase oxygen free-radical damage of the testicular microsomal P-450. Steroids 25-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-49 3795953-8 1986 The results suggest that steroid binding to P-450 is necessary but not sufficient to increase oxygen free-radical damage of the testicular microsomal P-450. Steroids 25-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-155 3795953-8 1986 The results suggest that steroid binding to P-450 is necessary but not sufficient to increase oxygen free-radical damage of the testicular microsomal P-450. Oxygen 94-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-49 3795953-9 1986 Testosterone acetate, which diminishes P-450 loss in Leydig cell cultures, was also bound stoichiometrically with high affinity (Kd = 17 nM) and produced a unique spectra, with maxima at 408 nm, an isosbestic point at 418 nm and minima at 428 nm. testosterone acetate 0-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-44 2424910-0 1986 The cDNA and protein sequence of a phenobarbital-induced chicken cytochrome P-450. Phenobarbital 35-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-81 3741479-0 1986 Inhibition of aromatase cytochrome P-450 (estrogen synthetase) by derivatives of alpha-naphthoflavone. alpha-naphthoflavone 81-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-40 3741479-1 1986 alpha-Naphthoflavone (ANF; 7,8-benzoflavone) is a potent competitive inhibitor of human aromatase cytochrome P-450 [J. T. Kellis, Jr. and L. E. Vickery, Science 225, 1032 (1984)]. alpha-naphthoflavone 0-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-114 3741479-1 1986 alpha-Naphthoflavone (ANF; 7,8-benzoflavone) is a potent competitive inhibitor of human aromatase cytochrome P-450 [J. T. Kellis, Jr. and L. E. Vickery, Science 225, 1032 (1984)]. alpha-naphthoflavone 22-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-114 3741479-1 1986 alpha-Naphthoflavone (ANF; 7,8-benzoflavone) is a potent competitive inhibitor of human aromatase cytochrome P-450 [J. T. Kellis, Jr. and L. E. Vickery, Science 225, 1032 (1984)]. alpha-naphthoflavone 27-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-114 3741479-8 1986 9-Hydroxy-ANF also induced a change in the absorption spectrum of hte aromatase cytochrome P-450 indicative of substrate displacement. 9-hydroxy-alpha-naphthoflavone 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-96 3022748-4 1986 This decrease at high P-450/reductase ratios could reflect tight coupling of reductase to P-450-PB, therefore decreasing electron transfer from reductase to ferric-EDTA, or could involve non-specific scavenging of .OH by P-450-PB. Fe(III)-EDTA 157-168 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-37 3746811-0 1986 Oxidation of 4-aryl- and 4-alkyl-substituted 2,6-dimethyl-3,5-bis(alkoxycarbonyl)-1,4-dihydropyridines by human liver microsomes and immunochemical evidence for the involvement of a form of cytochrome P-450. 4-aryl- and 4-alkyl-substituted 2,6-dimethyl-3,5-bis(alkoxycarbonyl)-1,4-dihydropyridines 13-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 201-206 3746811-4 1986 The 4-alkyl compounds, in contrast, formed a pyridine derivative in which a hydrogen atom was present at the 4-position and the alkyl group was lost; these compounds also inactivated cytochrome P-450 and caused the loss of nifedipine oxidase activity after enzymatic oxidation. alkyl 6-11 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 194-199 3746811-4 1986 The 4-alkyl compounds, in contrast, formed a pyridine derivative in which a hydrogen atom was present at the 4-position and the alkyl group was lost; these compounds also inactivated cytochrome P-450 and caused the loss of nifedipine oxidase activity after enzymatic oxidation. pyridine 45-53 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 194-199 3746811-4 1986 The 4-alkyl compounds, in contrast, formed a pyridine derivative in which a hydrogen atom was present at the 4-position and the alkyl group was lost; these compounds also inactivated cytochrome P-450 and caused the loss of nifedipine oxidase activity after enzymatic oxidation. Hydrogen 76-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 194-199 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 2 subfamily B member 6 Homo sapiens 63-70 3746811-7 1986 The results indicate that P-450NF (or closely related enzyme forms) is responsible for the oxidation of these nifedipine-related compounds in human liver microsomes and that metabolism is highly dependent upon 4-substitution; with alkyl substituents, radicals are postulated to leave P-450NF to attack other proteins. Nifedipine 110-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-33 3746811-7 1986 The results indicate that P-450NF (or closely related enzyme forms) is responsible for the oxidation of these nifedipine-related compounds in human liver microsomes and that metabolism is highly dependent upon 4-substitution; with alkyl substituents, radicals are postulated to leave P-450NF to attack other proteins. Nifedipine 110-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 284-291 2424910-1 1986 Several cDNA clones complementary to a chicken phenobarbital-inducible cytochrome P-450 have been isolated and sequenced, representing the first non-mammalian eukaryotic cytochrome P-450 sequence to be analyzed. Phenobarbital 47-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-87 2424910-1 1986 Several cDNA clones complementary to a chicken phenobarbital-inducible cytochrome P-450 have been isolated and sequenced, representing the first non-mammalian eukaryotic cytochrome P-450 sequence to be analyzed. Phenobarbital 47-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 181-186 2424910-6 1986 The chicken cytochrome P-450 shows an overall homology of 45-54% compared with the mammalian phenobarbital-induced cytochrome P-450s. Phenobarbital 93-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 23-28 2424910-8 1986 In particular, the chicken cytochrome P-450 contains the conserved cysteinyl domain near the carboxyl terminus, found in all cytochrome P-450s and which is thought to be involved in heme binding. Heme 182-186 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 38-43 2424910-10 1986 It is suggested that the phenobarbital-, 3-methylcholanthrene, and pregnenolone 16 alpha-carbonitrile-induced cytochrome P-450 gene families diverged from a common ancestral gene 600 million years ago. phenobarbital-, 3-methylcholanthrene 25-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-126 2424910-10 1986 It is suggested that the phenobarbital-, 3-methylcholanthrene, and pregnenolone 16 alpha-carbonitrile-induced cytochrome P-450 gene families diverged from a common ancestral gene 600 million years ago. Pregnenolone 67-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-126 2424910-10 1986 It is suggested that the phenobarbital-, 3-methylcholanthrene, and pregnenolone 16 alpha-carbonitrile-induced cytochrome P-450 gene families diverged from a common ancestral gene 600 million years ago. alpha-carbonitrile 83-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-126 3087972-4 1986 We now report that digitoxigenin monodigitoxoside UDP-glucuronosyltransferase (DIG UDP-glucuronosyltransferase), a liver microsomal enzyme activity induced by PCN in rats, is also inducible, as is P-450p, in primary monolayer cultures of adult rat hepatocytes. Digitoxigenin 19-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 197-203 3087972-4 1986 We now report that digitoxigenin monodigitoxoside UDP-glucuronosyltransferase (DIG UDP-glucuronosyltransferase), a liver microsomal enzyme activity induced by PCN in rats, is also inducible, as is P-450p, in primary monolayer cultures of adult rat hepatocytes. Pregnenolone Carbonitrile 159-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 197-203 3087972-7 1986 We conclude that the liver enzymes controlled by the postulated PCN recognition system include not only P-450p but also one or more UDP-glucuronosyltransferases. Pregnenolone Carbonitrile 64-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-110 3702869-0 1986 [Comparison of the heme electron state of reduced cytochrome P450 and P420 in equilibrium and non-equilibrium protein conformations. Heme 19-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-65 3702869-4 1986 The comparison of the Soret MCD spectra of reduced proteins in their equilibrium and non-equilibrium forms with those of other high-spin ferrous hemoproteins suggest that mercaptide (RS-) is the protein ligand of the heme iron in reduced P450, as well as in its CO-complex, and that imidazole of histidine is the fifth ligand of the iron both in reduced P420 and its CO-complex. mercaptide 171-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 238-242 3947346-2 1986 The enzyme converts androstenedione to estrone (Vmax 13.3 n moles/min/n mole P-450; Km 30 microM) and testosterone to estradiol. Androstenedione 20-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-82 3753972-0 1986 The insulin-like growth factor, somatomedin C, induces the synthesis of cholesterol side-chain cleavage cytochrome P-450 and adrenodoxin in ovarian cells. Cholesterol 72-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 115-120 3947346-2 1986 The enzyme converts androstenedione to estrone (Vmax 13.3 n moles/min/n mole P-450; Km 30 microM) and testosterone to estradiol. Estrone 39-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-82 3947346-2 1986 The enzyme converts androstenedione to estrone (Vmax 13.3 n moles/min/n mole P-450; Km 30 microM) and testosterone to estradiol. Testosterone 102-114 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-82 3947346-2 1986 The enzyme converts androstenedione to estrone (Vmax 13.3 n moles/min/n mole P-450; Km 30 microM) and testosterone to estradiol. Estradiol 118-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-82 3921402-5 1985 However, phenobarbital-inducible proteins were identified on "Western blots" using antibodies to a rat liver phenobarbital inducible P-450 form. Phenobarbital 9-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-138 3766260-0 1986 Further evidence for a role of isozymes of P450 in the metabolism and toxicity of carbon disulfide (CS2). Carbon Disulfide 82-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-47 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 2 subfamily B member 6 Homo sapiens 192-198 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 2 subfamily B member 6 Homo sapiens 192-198 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 2 subfamily B member 6 Homo sapiens 192-198 2986550-0 1985 Regulation of steroidogenesis in rat Leydig cells in culture: effect of human chorionic gonadotropin and dibutyryl cyclic AMP on the synthesis of cholesterol side chain cleavage cytochrome P-450 and adrenodoxin. Bucladesine 105-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-210 2986550-0 1985 Regulation of steroidogenesis in rat Leydig cells in culture: effect of human chorionic gonadotropin and dibutyryl cyclic AMP on the synthesis of cholesterol side chain cleavage cytochrome P-450 and adrenodoxin. Cholesterol 146-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-210 2986550-1 1985 Rat Leydig cells in primary culture were used as a model system to investigate the effects of human chorionic gonadotropin (hCG) and dibutyryl cyclic AMP (Bt2cAMP) on the synthesis of cholesterol side chain cleavage cytochrome P-450 (cytochrome P-450scc) and the iron-sulfur protein, adrenodoxin. Cholesterol 184-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 227-232 4006010-4 1985 Compounds possessing intact allyl and methylenedioxyphenyl groups (safrole, isosafrole and myristicine) were the most potent inducers of cytochromes P-450 and P-448; compounds containing an intact allyl group only (estragole, allybenzene and eugenol methyl ether) or an oxidized allyl group and an intact methylenedioxyphenyl group (epoxysafrole) were inducers of P-448 only. Safrole 67-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-164 4006010-4 1985 Compounds possessing intact allyl and methylenedioxyphenyl groups (safrole, isosafrole and myristicine) were the most potent inducers of cytochromes P-450 and P-448; compounds containing an intact allyl group only (estragole, allybenzene and eugenol methyl ether) or an oxidized allyl group and an intact methylenedioxyphenyl group (epoxysafrole) were inducers of P-448 only. isosafrole 76-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-164 4006010-4 1985 Compounds possessing intact allyl and methylenedioxyphenyl groups (safrole, isosafrole and myristicine) were the most potent inducers of cytochromes P-450 and P-448; compounds containing an intact allyl group only (estragole, allybenzene and eugenol methyl ether) or an oxidized allyl group and an intact methylenedioxyphenyl group (epoxysafrole) were inducers of P-448 only. myristicin 91-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-164 4039566-4 1985 When the observed rates of 7,8 and 9,10 diol formation per nmole P450 for chow-fed animals are plotted vs. the b5/P450 ratio a positive correlation was observed suggesting that cytochrome b5 participates directly in the microsomal metabolism of benzo(a)pyrene. Benzo(a)pyrene 245-259 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-118 3921402-5 1985 However, phenobarbital-inducible proteins were identified on "Western blots" using antibodies to a rat liver phenobarbital inducible P-450 form. Phenobarbital 109-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 133-138 3921402-7 1985 However, characteristic of P-450 forms, the synthesis of these proteins was suppressed by 3-methylcholanthrene treatment. Methylcholanthrene 90-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-32 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 2 subfamily B member 6 Homo sapiens 88-93 6525204-5 1984 The maximal synthetic rates of aldosterone and 18-hydroxycorticosterone were 2 and 5 nmol/nmol P-450/min, respectively. Aldosterone 31-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-104 6525204-5 1984 The maximal synthetic rates of aldosterone and 18-hydroxycorticosterone were 2 and 5 nmol/nmol P-450/min, respectively. 18-Hydroxycorticosterone 47-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-104 6489339-0 1984 Mechanism of rate control of the NADPH-dependent reduction of cytochrome P-450 by lipids in reconstituted phospholipid vesicles. NADP 33-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-78 3918581-1 1985 When Bacillus megaterium ATCC 14581 is grown in the presence of barbiturates, a cytochrome P-450-dependent fatty acid monooxygenase (Mr 120000) is induced (Kim, B.-H. and Fulco, A.J. Barbiturates 64-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 91-96 3918581-7 1985 Gel filtration chromatography of a crude monooxygenase preparation from pentobarbital-induced B. megaterium indicated that not all of the induced cytochrome P-450 present in the extract was accounted for by this high-molecular-weight component. Pentobarbital 72-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-162 4084271-0 1985 [Determination of caffeine and metamizole elimination in pregnancy and after delivery as an in vivo method for characterization of various cytochrome p-450 dependent biotransformation reactions]. Caffeine 18-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 150-155 6489339-0 1984 Mechanism of rate control of the NADPH-dependent reduction of cytochrome P-450 by lipids in reconstituted phospholipid vesicles. Phospholipids 106-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-78 6489339-1 1984 The NADPH-supported reduction of cytochrome P-450 LM2 (liver microsomal isozyme 2) in reconstituted phospholipid vesicles in general exhibits two-exponential kinetics. NADP 4-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-49 6489339-1 1984 The NADPH-supported reduction of cytochrome P-450 LM2 (liver microsomal isozyme 2) in reconstituted phospholipid vesicles in general exhibits two-exponential kinetics. Phospholipids 100-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 44-49 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 2 subfamily B member 6 Homo sapiens 95-100 6541651-3 1984 To help clarify this problem, we have carried out extensive studies of thiol-binding properties of native ferric chloroperoxidase and have compared our new results with those previously obtained in our laboratory on thiol adducts of P-450. Sulfhydryl Compounds 216-221 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 233-238 6541651-4 1984 We have found that the ligation of exogenous thiols to the heme iron of chloroperoxidase generates hyperporphyrin (split Soret) spectra (lambda max = approximately 372 and approximately 455 nm), consistent with the formation of bisthiolate low-spin ferric heme adducts as has been established for P-450 and its heme models. Sulfhydryl Compounds 45-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 297-302 6541651-4 1984 We have found that the ligation of exogenous thiols to the heme iron of chloroperoxidase generates hyperporphyrin (split Soret) spectra (lambda max = approximately 372 and approximately 455 nm), consistent with the formation of bisthiolate low-spin ferric heme adducts as has been established for P-450 and its heme models. Heme 59-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 297-302 6541651-4 1984 We have found that the ligation of exogenous thiols to the heme iron of chloroperoxidase generates hyperporphyrin (split Soret) spectra (lambda max = approximately 372 and approximately 455 nm), consistent with the formation of bisthiolate low-spin ferric heme adducts as has been established for P-450 and its heme models. Iron 64-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 297-302 6541651-4 1984 We have found that the ligation of exogenous thiols to the heme iron of chloroperoxidase generates hyperporphyrin (split Soret) spectra (lambda max = approximately 372 and approximately 455 nm), consistent with the formation of bisthiolate low-spin ferric heme adducts as has been established for P-450 and its heme models. hyperporphyrin 99-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 297-302 6696444-1 1984 Three alicyclic compounds (D-camphor, adamantanone, adamantane) were found to be hydroxylated by the cytochrome P-450 isoenzymes P-450cam and P-450LM2. alicyclic compounds 6-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-117 6474163-3 1984 7,8-Benzoflavone and chrysin were potent competitive inhibitors and induced spectral changes in the aromatase cytochrome P-450 indicative of substrate displacement. alpha-naphthoflavone 0-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-126 6474163-3 1984 7,8-Benzoflavone and chrysin were potent competitive inhibitors and induced spectral changes in the aromatase cytochrome P-450 indicative of substrate displacement. chrysin 21-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 121-126 6723577-8 1984 4- Cyclohexylaniline and d-aminoglutethimide were also assayed for their inhibition of cytochrome P-450-catalyzed cholesterol side-chain cleavage. 4-cyclohexylaniline 0-20 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-103 6723577-8 1984 4- Cyclohexylaniline and d-aminoglutethimide were also assayed for their inhibition of cytochrome P-450-catalyzed cholesterol side-chain cleavage. (S)-aminoglutethimide 25-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-103 6723577-8 1984 4- Cyclohexylaniline and d-aminoglutethimide were also assayed for their inhibition of cytochrome P-450-catalyzed cholesterol side-chain cleavage. Cholesterol 114-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-103 6723577-11 1984 Difference spectral measurements using crude placental microsomes and cholate extracts of these microsomes show that binding of 4- cyclohexylaniline produces a type II spectral change; this is indicative of coordination of the arylamine to the heme-iron of the aromatase cytochrome P-450. Cholates 70-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 282-287 6723577-11 1984 Difference spectral measurements using crude placental microsomes and cholate extracts of these microsomes show that binding of 4- cyclohexylaniline produces a type II spectral change; this is indicative of coordination of the arylamine to the heme-iron of the aromatase cytochrome P-450. 4-cyclohexylaniline 128-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 282-287 6723577-11 1984 Difference spectral measurements using crude placental microsomes and cholate extracts of these microsomes show that binding of 4- cyclohexylaniline produces a type II spectral change; this is indicative of coordination of the arylamine to the heme-iron of the aromatase cytochrome P-450. aniline 227-236 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 282-287 6723577-11 1984 Difference spectral measurements using crude placental microsomes and cholate extracts of these microsomes show that binding of 4- cyclohexylaniline produces a type II spectral change; this is indicative of coordination of the arylamine to the heme-iron of the aromatase cytochrome P-450. heme-iron 244-253 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 282-287 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 2 subfamily B member 6 Homo sapiens 11-16 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 2 subfamily B member 6 Homo sapiens 192-197 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 2 subfamily B member 6 Homo sapiens 11-16 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 2 subfamily B member 6 Homo sapiens 192-197 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 2 subfamily B member 6 Homo sapiens 11-16 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 2 subfamily B member 6 Homo sapiens 11-16 6438379-3 1984 Cytochrome P-450, NADPH-cytochrome P-450 reductase and NADPH were required for 5-FU production. Fluorouracil 79-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-16 6438379-4 1984 Inhibitors of cytochrome P-450 such as carbon monoxide and metyrapone markedly decreased the rate. Carbon Monoxide 39-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-30 6438379-4 1984 Inhibitors of cytochrome P-450 such as carbon monoxide and metyrapone markedly decreased the rate. Metyrapone 59-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 25-30 6696444-1 1984 Three alicyclic compounds (D-camphor, adamantanone, adamantane) were found to be hydroxylated by the cytochrome P-450 isoenzymes P-450cam and P-450LM2. Camphor 27-36 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-117 6696444-1 1984 Three alicyclic compounds (D-camphor, adamantanone, adamantane) were found to be hydroxylated by the cytochrome P-450 isoenzymes P-450cam and P-450LM2. adamantanone 38-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-117 6696444-1 1984 Three alicyclic compounds (D-camphor, adamantanone, adamantane) were found to be hydroxylated by the cytochrome P-450 isoenzymes P-450cam and P-450LM2. Adamantane 52-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-117 6679766-1 1983 The separation of the two domains, disclosed by limited trypsinolysis in the cholesterol side chain cleavage cytochrome P-450, by covalent chromatography on thiopropyl-Sepharose 6B is described. Cholesterol 77-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-125 6100256-1 1984 A rate-determining step in the cAMP-dependent action of ACTH on adrenal steroid biosynthesis is the interaction of cholesterol substrate with the cholesterol side-chain cleavage cytochrome P-450 in the mitochondrion. Cyclic AMP 31-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-194 6100256-1 1984 A rate-determining step in the cAMP-dependent action of ACTH on adrenal steroid biosynthesis is the interaction of cholesterol substrate with the cholesterol side-chain cleavage cytochrome P-450 in the mitochondrion. Steroids 72-79 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-194 6100256-1 1984 A rate-determining step in the cAMP-dependent action of ACTH on adrenal steroid biosynthesis is the interaction of cholesterol substrate with the cholesterol side-chain cleavage cytochrome P-450 in the mitochondrion. Cholesterol 115-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-194 6100256-1 1984 A rate-determining step in the cAMP-dependent action of ACTH on adrenal steroid biosynthesis is the interaction of cholesterol substrate with the cholesterol side-chain cleavage cytochrome P-450 in the mitochondrion. Cholesterol 146-157 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 189-194 6661254-0 1983 Direct in vitro effects of bis(tri-n-butyltin)oxide on hepatic cytochrome P-450. bis(tri-n-butyltin)oxide 27-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-79 6679766-0 1983 [Separation of cholesterol-specific cytochrome P-450 regions and their localization in the polypeptide chain]. Cholesterol 15-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-52 6679766-1 1983 The separation of the two domains, disclosed by limited trypsinolysis in the cholesterol side chain cleavage cytochrome P-450, by covalent chromatography on thiopropyl-Sepharose 6B is described. thiopropyl-sepharose 157-180 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-125 6643443-6 1983 Comparison of the MCD spectra of the second category to those of ferrous cytochromes b5, c, and P-420 suggests that the axial cysteinate ligand is still present in the nonhyper ferrous P-450 species. cysteinate 126-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 185-190 6643443-7 1983 Thus, the combination of a strongly electron-donating cysteinate ligand and a trans sigma-donor, not the orbital mixing mechanism, is most likely the origin of the red-shifted Soret absorption maximum of nonhyper ferrous P-450 ligand complexes. cysteinate 54-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 221-226 6639671-3 1983 Accordingly, acute dietary iron deprivation is found to result in a marked decrease in I-P-450 content and activity. Iron 27-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-94 6639671-5 1983 We investigated the mechanistic basis for such acute reduction and report that iron was not only required as a co-substrate for I-P-450 heme formation, but also as a regulator of two key heme-synthetic enzymes, delta-aminolevulinic acid synthetase and ferrochelatase. Iron 79-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-135 6639671-5 1983 We investigated the mechanistic basis for such acute reduction and report that iron was not only required as a co-substrate for I-P-450 heme formation, but also as a regulator of two key heme-synthetic enzymes, delta-aminolevulinic acid synthetase and ferrochelatase. Heme 136-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-135 6639671-6 1983 In addition, our studies revealed that dietary deprivation of selenium for a single day dramatically reduced I-P-450-dependent MFO activity. Selenium 62-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-116 6643443-8 1983 Further, the nature of the total electronic interactions between both axial ligands and the heme iron of ferrous P-450 and not solely the cysteinate ligand determines whether the ligand complexes will be of the hyper or nonhyperporphyrin category. Heme 92-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-118 6639671-7 1983 This prompt reduction apparently reflects impaired I-P-450 formation resulting from lowered ferrochelatase activity and consequently decreased intestinal heme availability, and was not a consequence of intracellular peroxidation presumably enhanced by concomitant lowering of the seleno-dependent glutathione peroxidase. Heme 154-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-58 6639671-8 1983 Thus, we report the novel observation that dietary selenium also appears to be a critical modulator of intestinal cytochrome P-450-dependent metabolism of ingested drugs, carcinogens, and toxins that are absorbed by the intestinal mucosa. Selenium 51-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 125-130 6643443-8 1983 Further, the nature of the total electronic interactions between both axial ligands and the heme iron of ferrous P-450 and not solely the cysteinate ligand determines whether the ligand complexes will be of the hyper or nonhyperporphyrin category. Iron 97-101 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-118 6643443-8 1983 Further, the nature of the total electronic interactions between both axial ligands and the heme iron of ferrous P-450 and not solely the cysteinate ligand determines whether the ligand complexes will be of the hyper or nonhyperporphyrin category. cysteinate 138-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 113-118 6870907-2 1983 Within a series of benzphetamine analogues the liver microsomal enzyme system exhibits a close correlation of the substrate induced spin equilibrium shift towards the high spin state and both the rate of P-450 reduction, and of substrate turnover, as well. Benzphetamine 19-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 204-209 7424328-0 1980 The influence of allyl isopropyl acetamide on d-aminolevulinic acid synthetase and cytochrome P-450. Allylisopropylacetamide 17-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-99 6303756-0 1983 Biosynthesis of cholesterol side-chain cleavage cytochrome P-450 in human fetal adrenal cells in culture. Cholesterol 16-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-64 6303756-1 1983 An immunoglobulin G fraction from antiserum raised against cholesterol side-chain cleavage cytochrome P-450 (cytochrome P-450sec) purified from bovine adrenocortical mitochondria cross-reacted with human fetal adrenal cytochrome P-450sec in an Ouchterlony double diffusion system. Cholesterol 59-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-107 6303756-1 1983 An immunoglobulin G fraction from antiserum raised against cholesterol side-chain cleavage cytochrome P-450 (cytochrome P-450sec) purified from bovine adrenocortical mitochondria cross-reacted with human fetal adrenal cytochrome P-450sec in an Ouchterlony double diffusion system. Cholesterol 59-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 120-128 6303756-1 1983 An immunoglobulin G fraction from antiserum raised against cholesterol side-chain cleavage cytochrome P-450 (cytochrome P-450sec) purified from bovine adrenocortical mitochondria cross-reacted with human fetal adrenal cytochrome P-450sec in an Ouchterlony double diffusion system. Cholesterol 59-70 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 229-237 6416251-3 1983 The NADPH-dependent reduction of P-450 LM2 has been studied both anaerobically and aerobically in solution state. NADP 4-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-38 6810958-1 1982 The low molecular weight analog of superoxide dismutase, th Cu(Lys)2 complex inhibits the oxidation of type I (piperidinoanthraquinone) and type II(aniline) substrates catalyzed by cytochrome P-450. 1-Piperidinoanthraquinone 111-134 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-197 6810958-1 1982 The low molecular weight analog of superoxide dismutase, th Cu(Lys)2 complex inhibits the oxidation of type I (piperidinoanthraquinone) and type II(aniline) substrates catalyzed by cytochrome P-450. aniline 148-155 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 192-197 6617929-5 1983 P-450 was significantly correlated with lactate (rs -0.79), pyruvate (rs -0.65) and serum insulin (rs -0.53). Lactic Acid 40-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 6617929-5 1983 P-450 was significantly correlated with lactate (rs -0.79), pyruvate (rs -0.65) and serum insulin (rs -0.53). Pyruvic Acid 60-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-5 6975411-6 1981 The formation of omega-hydroxydodecanol was more sharply inhibited by CO than was the formation of (omega-1)-hydroxydodecanol, implying that more than one cytochrome P-450 was involved in the hydroxylation of 1-dodecanol and that CO has a higher affinity for the P-450 catalyzing the omega-hydroxylation. omega-hydroxydodecanol 17-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-171 6975411-6 1981 The formation of omega-hydroxydodecanol was more sharply inhibited by CO than was the formation of (omega-1)-hydroxydodecanol, implying that more than one cytochrome P-450 was involved in the hydroxylation of 1-dodecanol and that CO has a higher affinity for the P-450 catalyzing the omega-hydroxylation. omega-hydroxydodecanol 17-39 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 263-268 6975411-6 1981 The formation of omega-hydroxydodecanol was more sharply inhibited by CO than was the formation of (omega-1)-hydroxydodecanol, implying that more than one cytochrome P-450 was involved in the hydroxylation of 1-dodecanol and that CO has a higher affinity for the P-450 catalyzing the omega-hydroxylation. (omega-1)-hydroxydodecanol 99-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-171 6975411-6 1981 The formation of omega-hydroxydodecanol was more sharply inhibited by CO than was the formation of (omega-1)-hydroxydodecanol, implying that more than one cytochrome P-450 was involved in the hydroxylation of 1-dodecanol and that CO has a higher affinity for the P-450 catalyzing the omega-hydroxylation. Dodecanol 209-220 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-171 7344454-0 1981 The heme protein P-450 in oxygen activation: carbon monoxide inhibition and photochemical action spectroscopy as tools to study its catalytic role. Oxygen 26-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-22 7344454-0 1981 The heme protein P-450 in oxygen activation: carbon monoxide inhibition and photochemical action spectroscopy as tools to study its catalytic role. Carbon Monoxide 45-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-22 7424328-1 1980 Allyl isopropyl acetamide (AIA) is believed to destroy P-450 and to enhance the d-aminolevulinic acid synthetase (ALAS) activity by lowering the heme pool of the liver parenchymal cell, in this way weakening the negative feed back mechanism, which regulates ALAS synthesis at the translational level. Allylisopropylacetamide 0-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 7424328-1 1980 Allyl isopropyl acetamide (AIA) is believed to destroy P-450 and to enhance the d-aminolevulinic acid synthetase (ALAS) activity by lowering the heme pool of the liver parenchymal cell, in this way weakening the negative feed back mechanism, which regulates ALAS synthesis at the translational level. Allylisopropylacetamide 27-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 55-60 118106-5 1979 Addition of phenobarbital or methylcholanthrene at day 5 in culture caused an increase in cytochromes P-450 and P-448, respectively, only in hepatocytes cultured on collagen membranes and confluent fibroblasts. Phenobarbital 12-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-117 227893-0 1979 Studies of the electron-nuclear coupling between Fe(III) and 14N in cytochrome P-450 and in a series of low spin heme compounds. ferric sulfate 49-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-84 227893-0 1979 Studies of the electron-nuclear coupling between Fe(III) and 14N in cytochrome P-450 and in a series of low spin heme compounds. 4-(4-methylpiperazin-1-yl)benzoic acid 61-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-84 227893-4 1979 We have found that the heme of low spin ferric cytochrome P-450 is coordinated to a nitrogenous ligand, probably imidazole. Heme 23-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-63 227893-4 1979 We have found that the heme of low spin ferric cytochrome P-450 is coordinated to a nitrogenous ligand, probably imidazole. imidazole 113-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 58-63 40703-0 1979 The interaction of aliphatic analogs of methylene-dioxyphenyl compounds with cytochromes P-450 and P-420. aliphatic 19-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-104 40703-0 1979 The interaction of aliphatic analogs of methylene-dioxyphenyl compounds with cytochromes P-450 and P-420. methylene-dioxyphenyl compounds 40-71 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 89-104 40703-1 1979 The spectra resulting from the interaction of a series of substituted dioxolanes with microsomal cytochromes P-450 or P-420, as well as purified cytochrome P-450, were measured. Dioxolanes 70-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-123 118106-5 1979 Addition of phenobarbital or methylcholanthrene at day 5 in culture caused an increase in cytochromes P-450 and P-448, respectively, only in hepatocytes cultured on collagen membranes and confluent fibroblasts. Methylcholanthrene 29-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-117 1061127-0 1976 Magnetic circular dichroism of ferrous carbonyl adducts of cytochromes P-450 and P-420 and their synthetic models: further evidence for mercaptide as the fifth ligand to iron. mercaptide 136-146 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-86 517009-5 1979 2) Triton N-101 provokes splitting of the associated structure both of solubilized P-450 and P-450 LM2; this effect is reversible. Triton N 101 3-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 83-102 28181-2 1978 All such compounds tested, with the exception of triphenyl phosphite, interact with ferrous cytochrome P-450 and its denatured form, cytochrome P-420, to produce complexes having two peaks in the Soret region of their optical difference spectra. triphenyl phosphite 49-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-108 356063-1 1978 The reviewed kinetic investigations at the experimentally accessible partial reactions of the reaction sequence clearly evidence that the P-450 system represents a complex reaction system, the main control points of which are shown in Scheme 3: As can be seen, control elements are the substrate, the cytochrome species (P-450/448), the reducing agent, the reductase, phospholipid and parts of the b5 system as well. Phospholipids 368-380 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 321-330 883972-0 1977 Kinetics of cumene hydroperoxide-dependent aniline hydroxylation involving cytochrome P-450 in microsomal and solubilized forms. cumene hydroperoxide 12-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-91 883972-0 1977 Kinetics of cumene hydroperoxide-dependent aniline hydroxylation involving cytochrome P-450 in microsomal and solubilized forms. aniline 43-50 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-91 976190-0 1976 Effects of spironolactone, canrenone and canrenoate-K on cytochrome P450, and 11beta- and 18-hydroxylation in bovine and human adrenal cortical mitochondria. Canrenoic Acid 41-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-72 976190-1 1976 Effects of spironolactone, canrenone and canrenoate-K on adrenal cytochrome P450 (P450) and corticosteroid biosynthesis were examined by studying difference spectra, P450 reduction and corticoid hydroxylation in mitochondrial preparations isolated from zona fasciculata and zona glomerulosa of bovine adrenals and from adrenal adenoma and hyperplastic adrenal cortex removed from patients with hyperaldosteronism. Spironolactone 11-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 976190-1 1976 Effects of spironolactone, canrenone and canrenoate-K on adrenal cytochrome P450 (P450) and corticosteroid biosynthesis were examined by studying difference spectra, P450 reduction and corticoid hydroxylation in mitochondrial preparations isolated from zona fasciculata and zona glomerulosa of bovine adrenals and from adrenal adenoma and hyperplastic adrenal cortex removed from patients with hyperaldosteronism. Spironolactone 11-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 976190-1 1976 Effects of spironolactone, canrenone and canrenoate-K on adrenal cytochrome P450 (P450) and corticosteroid biosynthesis were examined by studying difference spectra, P450 reduction and corticoid hydroxylation in mitochondrial preparations isolated from zona fasciculata and zona glomerulosa of bovine adrenals and from adrenal adenoma and hyperplastic adrenal cortex removed from patients with hyperaldosteronism. Spironolactone 11-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 976190-1 1976 Effects of spironolactone, canrenone and canrenoate-K on adrenal cytochrome P450 (P450) and corticosteroid biosynthesis were examined by studying difference spectra, P450 reduction and corticoid hydroxylation in mitochondrial preparations isolated from zona fasciculata and zona glomerulosa of bovine adrenals and from adrenal adenoma and hyperplastic adrenal cortex removed from patients with hyperaldosteronism. Canrenoic Acid 41-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 76-80 976190-1 1976 Effects of spironolactone, canrenone and canrenoate-K on adrenal cytochrome P450 (P450) and corticosteroid biosynthesis were examined by studying difference spectra, P450 reduction and corticoid hydroxylation in mitochondrial preparations isolated from zona fasciculata and zona glomerulosa of bovine adrenals and from adrenal adenoma and hyperplastic adrenal cortex removed from patients with hyperaldosteronism. Canrenoic Acid 41-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 976190-1 1976 Effects of spironolactone, canrenone and canrenoate-K on adrenal cytochrome P450 (P450) and corticosteroid biosynthesis were examined by studying difference spectra, P450 reduction and corticoid hydroxylation in mitochondrial preparations isolated from zona fasciculata and zona glomerulosa of bovine adrenals and from adrenal adenoma and hyperplastic adrenal cortex removed from patients with hyperaldosteronism. Canrenoic Acid 41-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-86 976190-5 1976 Spironolactone stimulated P450 reduction. Spironolactone 0-14 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-30 938524-0 1976 Ethanol-induced cytochrome P-450: catalytic activity after partial purification. Ethanol 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-32 1061127-2 1976 Ferrous porphyrins with sodium methyl mercaptide and CO in benzene give MCD and absorption spectra which are almost identical to those of the natural enzyme, indicating that in P-450 a mercaptide serves as the fifth ligand in the ferrous carbonyl adduct. Benzene 59-66 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-182 1061127-2 1976 Ferrous porphyrins with sodium methyl mercaptide and CO in benzene give MCD and absorption spectra which are almost identical to those of the natural enzyme, indicating that in P-450 a mercaptide serves as the fifth ligand in the ferrous carbonyl adduct. mercaptide 38-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-182 1061127-5 1976 The infrared stretching frequencies of ferrous porphyrin carbonyl complexes and the absorption spectrum of the CO adduct of Na[Fe1(meso-tetraphenylporphyrin dianion)] are consistent with the concept that in P-450 considerable electron density is transferred to the iron by the mercaptide ligand. Porphyrins 47-56 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 207-212 1061127-5 1976 The infrared stretching frequencies of ferrous porphyrin carbonyl complexes and the absorption spectrum of the CO adduct of Na[Fe1(meso-tetraphenylporphyrin dianion)] are consistent with the concept that in P-450 considerable electron density is transferred to the iron by the mercaptide ligand. fe1(meso-tetraphenylporphyrin dianion) 127-165 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 207-212 1061127-5 1976 The infrared stretching frequencies of ferrous porphyrin carbonyl complexes and the absorption spectrum of the CO adduct of Na[Fe1(meso-tetraphenylporphyrin dianion)] are consistent with the concept that in P-450 considerable electron density is transferred to the iron by the mercaptide ligand. Iron 265-269 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 207-212 1061127-5 1976 The infrared stretching frequencies of ferrous porphyrin carbonyl complexes and the absorption spectrum of the CO adduct of Na[Fe1(meso-tetraphenylporphyrin dianion)] are consistent with the concept that in P-450 considerable electron density is transferred to the iron by the mercaptide ligand. mercaptide 277-287 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 207-212 6968297-5 1980 Hexobarbital hydroxylating activity and binding capacity of P-450 for hexobarbital, which were strikingly decreased after alpha-irradiation were less protected by radical scavengers, in contrast to the case of the lipid peroxidation and of the enzymes. Hexobarbital 70-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-65 1270707-1 1976 Evidence for P-450 type heme environment from magnetic circular dichroism spectroscopy. Heme 24-28 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-18 1061127-2 1976 Ferrous porphyrins with sodium methyl mercaptide and CO in benzene give MCD and absorption spectra which are almost identical to those of the natural enzyme, indicating that in P-450 a mercaptide serves as the fifth ligand in the ferrous carbonyl adduct. ferroporphyrin 0-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-182 1061127-2 1976 Ferrous porphyrins with sodium methyl mercaptide and CO in benzene give MCD and absorption spectra which are almost identical to those of the natural enzyme, indicating that in P-450 a mercaptide serves as the fifth ligand in the ferrous carbonyl adduct. methylmercaptan 24-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 177-182 4150427-0 1973 The heme protein P-450 in steroid hydroxylation. Steroids 26-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-22 4156173-2 1974 The involvement of cytochrome b5 in the NADPH-supported cytochrome P-450-dependent hydroxylation of chlorobenzene. NADP 40-45 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-72 4156173-2 1974 The involvement of cytochrome b5 in the NADPH-supported cytochrome P-450-dependent hydroxylation of chlorobenzene. chlorobenzene 100-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-72 4412067-0 1974 Apparent conversion of placental cytochrome P-420 to P-450 with p-chloromercuribenzoate. p-chloromercuribenzoate 64-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-58 4387274-0 1968 Participation of the microsomal electron transport system involving cytochrome P-450 in omega-oxidation of fatty acids. Fatty Acids 107-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-84 4646802-0 1972 Damage effect of chronic intoxication by CCl 4 on structural organization of liver microsomes and cytochromes (b) 5 and P-450. Cefaclor 41-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 122-127 4348064-0 1972 Haem protein P-450 from the adrenal cortex: interaction with steroids and the hydroxylation reaction. Steroids 61-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-18 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 2 subfamily B member 6 Homo sapiens 192-207 4650614-1 1972 V. Competition between cytochromes P-450 and P-448 for reductase in 3,4-benzpyrene hydroxylation. Benzo(a)pyrene 68-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-64 5011965-0 1972 Substrate-induced difference spectra and cholesterol to pregnenolone conversion with adrenal heme protein P-450. Pregnenolone 56-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-111 6065969-0 1967 Effect of hexane and carbon tetrachloride on microsomal cytochrome (P450). Hexanes 10-16 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-72 6065969-0 1967 Effect of hexane and carbon tetrachloride on microsomal cytochrome (P450). Carbon Tetrachloride 21-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 56-72 4379553-0 1965 Effect of thyroxine on the level of cytochrome b5 and P450. Thyroxine 10-19 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-58 4292159-0 1967 Reconversion of detergent- and sulfhydryl reagent-produced P-420 to P-450 by polyols and glutathione. polyol 77-84 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-73 4292159-0 1967 Reconversion of detergent- and sulfhydryl reagent-produced P-420 to P-450 by polyols and glutathione. Glutathione 89-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-73 33652317-0 2021 Metabolic activation mechanism of 2,2",3,3",6,6"-hexachlorobiphenyl (PCB136) by cytochrome P450 2B6: A QM/MM approach. 2,3,6,2',3',6'-hexachlorobiphenyl 34-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-99 33652317-0 2021 Metabolic activation mechanism of 2,2",3,3",6,6"-hexachlorobiphenyl (PCB136) by cytochrome P450 2B6: A QM/MM approach. 2,3,6,2',3',6'-hexachlorobiphenyl 69-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 80-99 33652317-2 2021 The present work implemented quantum mechanic/molecular mechanic methods (QM/MM) and density functional theory (DFT) to study the metabolic activation of 2,2",3,3",6,6"-hexachlorobiphenyl (PCB136) catalyzed by CYP2B6. 2,3,6,2',3',6'-hexachlorobiphenyl 154-187 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 210-216 33652317-2 2021 The present work implemented quantum mechanic/molecular mechanic methods (QM/MM) and density functional theory (DFT) to study the metabolic activation of 2,2",3,3",6,6"-hexachlorobiphenyl (PCB136) catalyzed by CYP2B6. 2,3,6,2',3',6'-hexachlorobiphenyl 189-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 210-216 33895159-9 2021 In conclusion, ethanol downregulated miR-148a in hepatocytes through HNF4A regulation, which eventually decreased CYP2B6 expression. Ethanol 15-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 33336382-2 2021 Previous studies have shown that metamizole induces cytochrome (CYP) 2B6 and possibly CYP3A4. Dipyrone 33-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-72 33756427-7 2021 A common loci rs3181842 (high linkage equilibrium with rs2279345) in CYP2B6 at 19p13.2 were found to be strongly associated with low serum levels of p, p"-DDE in this population in GWAS and were verified by two replications and combined analysis of 2353 subjects (P = 1.00 x 10-22). Dichlorodiphenyl Dichloroethylene 149-158 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 33756427-9 2021 Furthermore, the rs3181842 C allele functionally led to low CYP2B6 expression and activity, resulting in a low metabolic capacity for the formation of p, p"-DDE from p, p"-DDT. Dichlorodiphenyl Dichloroethylene 151-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 33756427-9 2021 Furthermore, the rs3181842 C allele functionally led to low CYP2B6 expression and activity, resulting in a low metabolic capacity for the formation of p, p"-DDE from p, p"-DDT. DDT 166-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 33756427-10 2021 The study highlighted that CYP2B6 variants were more relevant than environmental exposure to internal p, p"-DDE exposure, which is important information for DDT risk assessments. Dichlorodiphenyl Dichloroethylene 102-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 33756427-10 2021 The study highlighted that CYP2B6 variants were more relevant than environmental exposure to internal p, p"-DDE exposure, which is important information for DDT risk assessments. DDT 157-160 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 33336382-9 2021 We confirmed these results in HepaRG cells, where 4-MAA, the principal metabolite of metamizole, induced the mRNA expression of CYP2B6, 2C9, 2C19 and 3A4. 4-maa 50-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-134 33336382-9 2021 We confirmed these results in HepaRG cells, where 4-MAA, the principal metabolite of metamizole, induced the mRNA expression of CYP2B6, 2C9, 2C19 and 3A4. Dipyrone 85-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-134 33336382-6 2021 In the clinical study, we confirmed a moderate induction of CYP2B6 (decrease in the efavirenz AUC by 79%) and 3A4 (decrease in the midazolam AUC by 68%) by metamizole. efavirenz 84-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 33336382-6 2021 In the clinical study, we confirmed a moderate induction of CYP2B6 (decrease in the efavirenz AUC by 79%) and 3A4 (decrease in the midazolam AUC by 68%) by metamizole. Midazolam 131-140 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 33336382-6 2021 In the clinical study, we confirmed a moderate induction of CYP2B6 (decrease in the efavirenz AUC by 79%) and 3A4 (decrease in the midazolam AUC by 68%) by metamizole. Dipyrone 156-166 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 33838175-0 2021 Increased toxicity and retention of Perflourooctane Sulfonate (PFOS) in humanized CYP2B6-Transgenic mice compared to Cyp2b-null mice is relieved by a High-Fat Diet (HFD). perflourooctane sulfonate 36-61 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 33872945-1 2021 Human hepatic cytochrome P450 2B6 (CYP2B6) expressed is responsible for the metabolism of many drugs, such as cyclophosphamide, ifosfamid, and efavirenz. Cyclophosphamide 110-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-33 33872945-1 2021 Human hepatic cytochrome P450 2B6 (CYP2B6) expressed is responsible for the metabolism of many drugs, such as cyclophosphamide, ifosfamid, and efavirenz. Cyclophosphamide 110-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 33872945-1 2021 Human hepatic cytochrome P450 2B6 (CYP2B6) expressed is responsible for the metabolism of many drugs, such as cyclophosphamide, ifosfamid, and efavirenz. Ifosfamide 128-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-33 33872945-1 2021 Human hepatic cytochrome P450 2B6 (CYP2B6) expressed is responsible for the metabolism of many drugs, such as cyclophosphamide, ifosfamid, and efavirenz. Ifosfamide 128-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 33872945-1 2021 Human hepatic cytochrome P450 2B6 (CYP2B6) expressed is responsible for the metabolism of many drugs, such as cyclophosphamide, ifosfamid, and efavirenz. efavirenz 143-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 14-33 33872945-1 2021 Human hepatic cytochrome P450 2B6 (CYP2B6) expressed is responsible for the metabolism of many drugs, such as cyclophosphamide, ifosfamid, and efavirenz. efavirenz 143-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 33838175-2 2021 Murine Cyp2b"s are induced by PFOS and high-fat diets (HFD) and therefore we hypothesized that human CYP2B6 may alleviate PFOS-induced steatosis. perfluorooctane sulfonic acid 30-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 33838175-2 2021 Murine Cyp2b"s are induced by PFOS and high-fat diets (HFD) and therefore we hypothesized that human CYP2B6 may alleviate PFOS-induced steatosis. perfluorooctane sulfonic acid 122-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 101-107 33838175-0 2021 Increased toxicity and retention of Perflourooctane Sulfonate (PFOS) in humanized CYP2B6-Transgenic mice compared to Cyp2b-null mice is relieved by a High-Fat Diet (HFD). perfluorooctane sulfonic acid 63-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 82-88 33838175-4 2021 Similar to murine Cyp2b10, CYP2B6 is inducible by PFOS. perfluorooctane sulfonic acid 50-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 27-33 34011532-1 2021 The squalene synthase inhibitor squalestatin 1 (Squal1) is a potent and efficacious inducer of CYP2B expression in primary cultured rat hepatocytes and rat liver. squalestatin 1 32-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-100 33838175-5 2021 Furthermore, three ND-fed hCYP2B6-Tg females treated with 10mg/kg/day PFOS died during the exposure period; neither Cyp2b-null nor HFD-fed mice died. perfluorooctane sulfonic acid 70-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-33 33838175-6 2021 hCYP2B6-Tg mice retained more PFOS in serum and liver than Cyp2b-null mice presumably causing the observed toxicity. perfluorooctane sulfonic acid 30-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-7 33838175-7 2021 In contrast, serum PFOS retention was reduced in the HFD-fed hCYP2B6-Tg mice; the opposite trend observed in HFD-fed Cyp2b-null mice. perfluorooctane sulfonic acid 19-23 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-68 33838175-9 2021 However, PFOS combined with a HFD exacerbated steatosis in all mice, especially in the hCYP2B6-Tg mice with significant disruption of key lipid metabolism genes such as Srebp1, Pparg, and Hmgcr. perfluorooctane sulfonic acid 9-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-94 33838175-10 2021 In conclusion, CYP2B6 is induced by PFOS but does not alleviate PFOS toxicity presumably due to increased retention. perfluorooctane sulfonic acid 36-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 33838175-11 2021 CYP2B6 protects from PFOS-mediated steatosis in ND-fed mice, but increases steatosis when co-treated with a HFD. perfluorooctane sulfonic acid 21-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 34011532-7 2021 Treatment with inhibitors of different steps of cholesterol biosynthesis attenuated CITCO-mediated CYP2B6 induction in HepaRG cells, and Prava treatment increased HMGCR expression and inhibited CYP2B6 induction with comparable potency. Cholesterol 48-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 34011532-7 2021 Treatment with inhibitors of different steps of cholesterol biosynthesis attenuated CITCO-mediated CYP2B6 induction in HepaRG cells, and Prava treatment increased HMGCR expression and inhibited CYP2B6 induction with comparable potency. Pravastatin 137-142 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 194-200 34011532-4 2021 However, treatment with farnesol, which mediates Squal1"s effect on rat CYP2B expression, increased CYP2B6 mRNA levels in HepaRG cells expressing the constitutive androstane receptor (CAR) but not in cells with knocked-down CAR. Farnesol 24-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-77 34011532-10 2021 Significance Statement The cholesterol biosynthesis inhibitor squalestatin 1 induces rat hepatic CYP2B expression indirectly, by causing accumulation of an endogenous isoprenoid that activates the constitutive androstane receptor (CAR). Cholesterol 27-38 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-102 34011532-10 2021 Significance Statement The cholesterol biosynthesis inhibitor squalestatin 1 induces rat hepatic CYP2B expression indirectly, by causing accumulation of an endogenous isoprenoid that activates the constitutive androstane receptor (CAR). squalestatin 1 62-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-102 34011532-6 2021 Prava treatment abolished CITCO-inducible CYP2B6 expression, but had less effect on rifampicin-mediated CYP3A4 induction, and CITCO treatment did not affect Prava-inducible HMG-CoA reductase (HMGCR) expression. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 26-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 42-48 34011532-10 2021 Significance Statement The cholesterol biosynthesis inhibitor squalestatin 1 induces rat hepatic CYP2B expression indirectly, by causing accumulation of an endogenous isoprenoid that activates the constitutive androstane receptor (CAR). Terpenes 167-177 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-102 32970826-8 2021 Recent studies have shown clinically significant increases in exposure to CYP2C8 substrates (repaglinide, dasabuvir, desloratidine) and a CYP2B6 substrate (s-sibutramine) following co-administration with clopidogrel, indicating that therapeutic strategies with clopidogrel should avoid these drugs. sibutramine 156-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 32667979-2 2021 In a Phase 1 dolutegravir-efavirenz interaction study, mean dolutegravir Cmin decreased by 60% and 85% among CYP2B6 normal and slow/intermediate metabolizers, respectively. dolutegravir 13-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 32667979-2 2021 In a Phase 1 dolutegravir-efavirenz interaction study, mean dolutegravir Cmin decreased by 60% and 85% among CYP2B6 normal and slow/intermediate metabolizers, respectively. efavirenz 26-35 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 32667979-2 2021 In a Phase 1 dolutegravir-efavirenz interaction study, mean dolutegravir Cmin decreased by 60% and 85% among CYP2B6 normal and slow/intermediate metabolizers, respectively. dolutegravir cmin 60-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 109-115 32970826-8 2021 Recent studies have shown clinically significant increases in exposure to CYP2C8 substrates (repaglinide, dasabuvir, desloratidine) and a CYP2B6 substrate (s-sibutramine) following co-administration with clopidogrel, indicating that therapeutic strategies with clopidogrel should avoid these drugs. Clopidogrel 204-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 32970826-8 2021 Recent studies have shown clinically significant increases in exposure to CYP2C8 substrates (repaglinide, dasabuvir, desloratidine) and a CYP2B6 substrate (s-sibutramine) following co-administration with clopidogrel, indicating that therapeutic strategies with clopidogrel should avoid these drugs. Clopidogrel 261-272 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 138-144 33995083-11 2021 Further studies should be conducted to confirm the role of SNPs in CYP2B6, CYP2D6, CYP1A2 and ABCB1 on the pharmacokinetic variability of dexketoprofen. dexketoprofen trometamol 138-151 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 67-73 33610566-8 2021 Moreover, isobavachalcone demonstrated broad-spectrum inhibitory effects against CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, UGT1A1, UGT1A9, UGT2B7 with IC50 values of 1.08-9.78 muM. isobavachalcone 10-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 33622176-2 2021 EST73502 is a new chemical entity intended for oral pain treatment with dual sigma-1 receptor (sigma1R) antagonism and mu-opioid receptor (MOR) partial agonism, that presents a promising potent analgesic activity.Several enzymes were involved in EST73502 metabolism catalysing the formation of different metabolites, CYP3A4 and CYP2D6 being the main ones.Fraction unbound was determined due to its impact in interactions, a considerable proportion of EST73502 being available.EST73502 showed a low potential for CYP inhibition, except for CYP2D6 that showed time-dependent inhibition.No induction potential was found for CYP1A2 and 3A4, while CYP2B6 was induced at high concentration.EST73502 seemed to be a potential efflux transporter substrate (efflux ratio >= 2) but a negligible in vivo impact would be expected due to its high solubility and permeability in Caco-2 cells. EST73502 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 643-649 32909316-6 2021 Isoniazid decreased efavirenz clearance by 7% in CYP2B6 normal metabolizers and 13% in slow and intermediate metabolizers. Isoniazid 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 33663326-2 2021 To determine the drug-drug interaction risk for cytochrome P450, and SLC and ABC transporters, brexpiprazole and its metabolite, DM-3411 were assessed in this in vitro investigation.Brexpiprazole exhibited weak inhibitory effects (IC50 >13 mumol/L) on CYP2C9, CYP2C19, CYP2D6 and CYP3A4 activities, but had moderate inhibitor activity on CYP2B6 (IC50 8.19 mumol/L). brexpiprazole 182-195 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 338-344 33395575-6 2021 RESULTS: The methanolic and ethyl acetate extracts inhibited CYP2B6 with IC50s 79.16 and 57.96 mug/ml respectively, while none of the extracts had any effect on rifampicin metabolism or showed time-dependant inhibition (TDI). ethyl acetate 28-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 61-67 33890704-6 2021 Treatment with metamizole, a CAR-dependent inducer of CYP2B6, CYP2C9, CYP2C19 and CYP3A4, accentuated the effect of deglucuronidation on AUC and MR. Dipyrone 15-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 54-60 33875422-10 2021 Between one in three and one in six individuals harbored reduced activity alleles of CYP2A6, CYP2B6, CYP2D6 and CYP2C8 with important implications for artemisinin, chloroquine and amodiaquine therapy. artemisinin 151-162 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 33875422-10 2021 Between one in three and one in six individuals harbored reduced activity alleles of CYP2A6, CYP2B6, CYP2D6 and CYP2C8 with important implications for artemisinin, chloroquine and amodiaquine therapy. Chloroquine 164-175 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 33875422-10 2021 Between one in three and one in six individuals harbored reduced activity alleles of CYP2A6, CYP2B6, CYP2D6 and CYP2C8 with important implications for artemisinin, chloroquine and amodiaquine therapy. Amodiaquine 180-191 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 93-99 32841585-6 2021 In terms of metabolism biomarkers, a lower methadone requirement was related to carriers of CYP2B6 genotypes *4(G/G) and *9(T/T) among Jewish patients, CYP2B6*9 genotype (T/T) and haplotypes (TA/TG); and CYP2C19 (*2/*2,*2/*3, and *3/*3; Han Chinese). Methadone 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 92-98 32841585-6 2021 In terms of metabolism biomarkers, a lower methadone requirement was related to carriers of CYP2B6 genotypes *4(G/G) and *9(T/T) among Jewish patients, CYP2B6*9 genotype (T/T) and haplotypes (TA/TG); and CYP2C19 (*2/*2,*2/*3, and *3/*3; Han Chinese). Methadone 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 152-158 32841585-8 2021 Lower methadone levels were reported in CYP2B6 SNPs, haplotypes TTT, and AGATAA (Han Chinese), CYP2C19 genotype *1/*1 (Han Chinese), allelic carrier *1xN (Caucasian), and CYP3A4 genotype *1/*1 (Caucasian). Methadone 6-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 32841585-9 2021 Carriers of CYP2B6 genotype *6/*6 (Caucasian), CYP2B6 haplotypes ATGCAG and ATGCTG (Han Chinese), and CYP3A4 genotype *1/*1B (Caucasian) had predicted higher methadone plasma levels. Methadone 158-167 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 33592181-6 2021 Cells expressing other CYPs had an even stronger effect, with those expressing CYP2B6, CYP5A1, CYP2A13, CYP51A1, or CYP1A2, respectively, being the strongest producers of O2- . Oxygen 171-174 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 33992954-0 2021 Plausible drug interaction between cyclophosphamide and voriconazole via inhibition of CYP2B6. Cyclophosphamide 35-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 33992954-0 2021 Plausible drug interaction between cyclophosphamide and voriconazole via inhibition of CYP2B6. Voriconazole 56-68 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-93 33992954-2 2021 Potent inhibition of CYP2B6 was noticeable for some azoles, including voriconazole. Azoles 52-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 33992954-2 2021 Potent inhibition of CYP2B6 was noticeable for some azoles, including voriconazole. Voriconazole 70-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 33992954-5 2021 Surveys of adverse event reporting databases in Japan (JADER) and the U.S. (FAERS) showed that the proportional reporting ratios of neutropenia, hemorrhagic cystitis, and alopecia for cyclophosphamide, which is principally activated by CYP2B6 in humans, were mostly reduced, or tended to be reduced when azoles, including voriconazole, were prescribed in combination. Cyclophosphamide 184-200 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 236-242 33739838-4 2021 Protein expression and intracellular unbound drug concentration (Kpuu) effects on the CLint were investigated for five prototypical probe substrates (bupropion-CYP2B6, diclofenac-CYP2C9, omeprazole-CYP2C19, bufuralol-CYP2D6, and midazolam-CYP3A4). Bupropion 150-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 160-166 32909316-6 2021 Isoniazid decreased efavirenz clearance by 7% in CYP2B6 normal metabolizers and 13% in slow and intermediate metabolizers. efavirenz 20-29 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 49-55 33546554-0 2021 Investigating the CYP2B6 rs3745274 and rs3211371 polymorphisms in Methadone-Responder and Non-Responder Addicts in Iran. Methadone 66-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 33599403-2 2021 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. artemisinin 43-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 33599403-2 2021 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. Bupropion 56-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 33599403-2 2021 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. Cyclophosphamide 67-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 33599403-2 2021 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. efavirenz 85-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 33599403-2 2021 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. Ketamine 96-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 33599403-2 2021 Drugs metabolized mainly by CYP2B6 include artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone. Methadone 110-119 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 33671323-0 2021 Pharmacokinetics of the CYP3A4 and CYP2B6 Inducer Carbamazepine and Its Drug-Drug Interaction Potential: A Physiologically Based Pharmacokinetic Modeling Approach. Carbamazepine 50-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 33671323-1 2021 The anticonvulsant carbamazepine is frequently used in the long-term therapy of epilepsy and is a known substrate and inducer of cytochrome P450 (CYP) 3A4 and CYP2B6. Carbamazepine 19-32 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 159-165 33671323-5 2021 The carbamazepine model applies metabolism by CYP3A4 and CYP2C8 to produce carbamazepine-10,11-epoxide, metabolism by CYP2B6 and UDP-glucuronosyltransferase (UGT) 2B7 and glomerular filtration. Carbamazepine 4-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 33833550-0 2021 Erratum: Association of CYP2B6 Genetic Variation with Efavirenz and Nevirapine Drug Resistance in HIV-1 Patients from Botswana [Corrigendum]. efavirenz 54-63 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 33833550-0 2021 Erratum: Association of CYP2B6 Genetic Variation with Efavirenz and Nevirapine Drug Resistance in HIV-1 Patients from Botswana [Corrigendum]. Nevirapine 68-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 33758532-0 2021 Association of CYP2B6 Genetic Variation with Efavirenz and Nevirapine Drug Resistance in HIV-1 Patients from Botswana. efavirenz 45-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 33758532-0 2021 Association of CYP2B6 Genetic Variation with Efavirenz and Nevirapine Drug Resistance in HIV-1 Patients from Botswana. Nevirapine 59-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 15-21 33758532-1 2021 Purpose: CYP2B6 liver enzyme metabolizes the two non-nucleoside reverse transcriptase inhibitors Efavirenz (EFV) and Nevirapine (NVP) used in the antiretroviral therapy (ART) regimens for HIV-infected individuals. efavirenz 97-106 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 33758532-1 2021 Purpose: CYP2B6 liver enzyme metabolizes the two non-nucleoside reverse transcriptase inhibitors Efavirenz (EFV) and Nevirapine (NVP) used in the antiretroviral therapy (ART) regimens for HIV-infected individuals. efavirenz 108-111 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 33758532-1 2021 Purpose: CYP2B6 liver enzyme metabolizes the two non-nucleoside reverse transcriptase inhibitors Efavirenz (EFV) and Nevirapine (NVP) used in the antiretroviral therapy (ART) regimens for HIV-infected individuals. Nevirapine 117-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 33758532-1 2021 Purpose: CYP2B6 liver enzyme metabolizes the two non-nucleoside reverse transcriptase inhibitors Efavirenz (EFV) and Nevirapine (NVP) used in the antiretroviral therapy (ART) regimens for HIV-infected individuals. Nevirapine 129-132 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 9-15 32652557-9 2021 CONCLUSION: Our findings support preliminary evidence that metamizole acts as a potent inductor of cytochrome P450 isoenzymes CYP2B6 and CYP3A4. Dipyrone 59-69 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 126-132 32648334-0 2021 Characterization of the Effect of Upadacitinib on the Pharmacokinetics of Bupropion, a Sensitive Cytochrome P450 2B6 Probe Substrate. upadacitinib 34-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-116 32648334-0 2021 Characterization of the Effect of Upadacitinib on the Pharmacokinetics of Bupropion, a Sensitive Cytochrome P450 2B6 Probe Substrate. Bupropion 74-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-116 32648334-1 2021 This phase 1 study characterized the effect of multiple doses of upadacitinib, an oral Janus kinase 1 selective inhibitor, on the pharmacokinetics of the cytochrome P450 (CYP) 2B6 substrate bupropion. upadacitinib 65-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-179 32648334-1 2021 This phase 1 study characterized the effect of multiple doses of upadacitinib, an oral Janus kinase 1 selective inhibitor, on the pharmacokinetics of the cytochrome P450 (CYP) 2B6 substrate bupropion. Bupropion 190-199 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-179 33622223-10 2021 Letermovir inhibited CYP2B6, CYP2C8, CYP3A, and UGT1A1 in vitro, and induced CYP3A4 and CYP2B6 in hepatocytes. letermovir 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 33622223-10 2021 Letermovir inhibited CYP2B6, CYP2C8, CYP3A, and UGT1A1 in vitro, and induced CYP3A4 and CYP2B6 in hepatocytes. letermovir 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-94 33546554-3 2021 Therefore, this study designed to examine the frequency of two SNPs of the CYP2B6 gene (rs3745274 and rs3211371) in addicted cases in two methadone-responders and methadone non-responders groups. Methadone 138-147 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 33546554-3 2021 Therefore, this study designed to examine the frequency of two SNPs of the CYP2B6 gene (rs3745274 and rs3211371) in addicted cases in two methadone-responders and methadone non-responders groups. Methadone 163-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 33546554-7 2021 Conclusion: Based on our findings, we can conclude that rs3745274 variant of CYP2B6 gene could serve as a potential biomarker, to evaluate the prognosis of addicted people fate under treatment with methadone. Methadone 198-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 32772362-4 2021 CYP2B6 activity in 72 healthy volunteers was determined using the disposition of efavirez as a probe drug. efavirez 81-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 33551819-6 2020 As expected, CITCO, the direct activator, and PB, the indirect activator of CAR, induced CYP3A4 (31 and 40-fold), CYP2B6 (24 and 28-fold) and UGT1A1 (2.9 and 4.2-fold), respectively. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 13-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 33572963-3 2021 CYP2B6, 3A4, 2C8, and 2C9 mRNA levels increased in a concentration-dependent manner in the presence of calcitriol in human cryopreserved hepatocytes and HepaRG cells. Calcitriol 103-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 33551819-6 2020 As expected, CITCO, the direct activator, and PB, the indirect activator of CAR, induced CYP3A4 (31 and 40-fold), CYP2B6 (24 and 28-fold) and UGT1A1 (2.9 and 4.2-fold), respectively. Phenobarbital 46-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 114-120 33176055-9 2021 Screening for putative substrates revealed binding of phenolic compounds such as L-mimosine and emodin, suggesting a potential application of this new thermophilic P450 in the production of the corresponding hydroxylated products. Mimosine 81-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-168 33087449-8 2021 Although the underlying mechanism of the down-regulation for CYP1A2 remain unclear, the presently reported results suggest that the down-regulation of CYP2B6 and CYP3A4 via HIF-alpha stabilization is caused by a decrease in the expressions of CAR, PXR, and RXR. -alpha 176-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 33176055-9 2021 Screening for putative substrates revealed binding of phenolic compounds such as L-mimosine and emodin, suggesting a potential application of this new thermophilic P450 in the production of the corresponding hydroxylated products. Emodin 96-102 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 164-168 33249052-9 2021 Mixtures containing BP caused a strong decrease of CAR transactivation in line with lower CYP2B6 expression. Benzo(a)pyrene 20-22 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 33355828-1 2020 Background: Africans exhibit great diversity in cytochrome P450 2B6 isoenzyme (CYP2B6), the major enzyme in efavirenz metabolism. efavirenz 108-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 48-77 32815870-8 2021 CYP2B6 poor metabolizers had greater efavirenz concentrations at all weeks and greater nevirapine concentrations at baseline. efavirenz 37-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32815870-8 2021 CYP2B6 poor metabolizers had greater efavirenz concentrations at all weeks and greater nevirapine concentrations at baseline. Nevirapine 87-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32750744-11 2020 Moreover, isobavachin exhibited broad inhibition against CYP2B6, 2C9, 2C19, UGT1A1, 1A9, 2B7 with Ki values from 0.05 to 3.05 mum. isobavachin 10-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 32870067-2 2020 The metabolism of methadone is complex and executed mainly by the cytochromes, CYP3A4 and CYP2B6. Methadone 18-27 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 32475982-1 2020 Bupropion is hydroxylated to its primary active metabolite hydroxybupropion by cytochrome P450 enzyme CYP2B6. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 32475982-1 2020 Bupropion is hydroxylated to its primary active metabolite hydroxybupropion by cytochrome P450 enzyme CYP2B6. hydroxybupropion 59-75 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 33355828-1 2020 Background: Africans exhibit great diversity in cytochrome P450 2B6 isoenzyme (CYP2B6), the major enzyme in efavirenz metabolism. efavirenz 108-117 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 79-85 33355828-2 2020 Aim: We examined the frequency of two functional single nucleotide polymorphisms (SNPs) of the CYP2B6 pharmacogene in HIV-infected Nigerians on efavirenz-based antiretroviral therapy. efavirenz 144-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-101 33112607-10 2020 CYP2B6, 2C9, and 2C19 are the main isoforms involved in temephos metabolism, and CYP3A4 and 2D6 have minor contributions. Temefos 56-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 33329709-0 2020 Effects of Genetic Polymorphisms of Drug Transporter ABCB1 (MDR1) and Cytochrome P450 Enzymes CYP2A6, CYP2B6 on Nicotine Addiction and Smoking Cessation. Nicotine 112-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 102-108 33255185-8 2020 Diazepam also promotes up-regulation of CYP2B6 in PHHs and in HepaRG cells. Diazepam 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 40-46 33060725-0 2020 Effects of CYP2B6 polymorphisms on plasma nevirapine concentrations: a systematic review and meta-analysis. Nevirapine 42-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 11-17 33124517-0 2020 Nonresponse to high-dose bupropion for depression in a patient carrying CYP2B6*6 and CYP2C19*17 variants: a case report. Bupropion 25-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 33124517-2 2020 Pharmacogenetic panel testing identified the patient as a carrier of the CYP2B6*6 allele, which has been associated with reduced bupropion metabolism and decreased concentrations of the pharmacologically active metabolite hydroxybupropion. Bupropion 129-138 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 33124517-2 2020 Pharmacogenetic panel testing identified the patient as a carrier of the CYP2B6*6 allele, which has been associated with reduced bupropion metabolism and decreased concentrations of the pharmacologically active metabolite hydroxybupropion. hydroxybupropion 222-238 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 33124517-4 2020 We propose a combined effect of the detected CYP2C19 and CYP2B6 genetic variants on bupropion metabolism. Bupropion 84-93 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 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 2 subfamily B member 6 Homo sapiens 85-91 33060725-7 2020 We found that carriers of the CYP2B6 516TT genotype had a 2.18 microg/mL higher NVP concentration than did the GG or GT (95% CI 1.28-3.08). Nevirapine 80-83 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 30-36 31766967-0 2020 microRNA-605 rs2043556 polymorphisms affect clopidogrel therapy through modulation of CYP2B6 and P2RY12 in acute coronary syndrome patients. clopidogrel 44-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 31766967-7 2020 The results demonstrate that miR-605 targets the mRNA of the CYP2B6 and P2RY12 genes, and that rs2043556 A/G polymorphisms in miR-605 modulate the mRNA and protein expression of CYP2B6 and P2RY12 differently, which may impact the effect of clopidogrel in ACS patients. clopidogrel 240-251 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 178-184 32320483-7 2020 Rifampicin requires solely PXR to induce CYP3A4 and CYP2B6, while phenobarbital-mediated induction of these CYPs did not show absolute dependency on either PXR or CAR suggesting its ability to switch nuclear receptor activation. Rifampin 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 32583524-9 2020 Orbitazine had no significant inhibitory effect on CYP1A2, CYP2B6, CYP2C9, or CYP2C19 in human liver microsomes, but showed a dose-dependent inhibition of CYP2C8, CYP2D6 and CYP3A4; and there was no orbitazine-mediated induction of CYP1A2, CYP2B6, CYP3A4 or mRNA expression in hepatocytes. orbitazine 0-10 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 240-246 32681429-4 2020 It is known that several androgen-metabolizing P450s (CYP3A4/5/43 and CYP2B6) and P450 enzymes (CYP2R1, CYP27A1, CYP27B1, CYP24A1, CYP3A4, CYP2J2), which are necessary for vitamin D metabolism, are expressed in the prostate. Vitamin D 172-181 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 32945596-1 2020 Ibrutinib may inhibit intestinal CYP3A4 and induce CYP2B6 and/or CYP3A. ibrutinib 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 51-57 32945596-3 2020 This phase I study evaluated the effect of ibrutinib on the pharmacokinetics of the CYP2B6 substrate bupropion, CYP3A substrate midazolam, and OCs ethinylestradiol (EE) and levonorgestrel (LN). ibrutinib 43-52 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 84-90 33008144-3 2020 CYP2D6 metabolizes the conversion of MAP to amphetamine (AMP), while CYP2B6 and CYP3A4 predominantly mediate the conversion of DMS to AMP. desmethylselegiline 127-130 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 33008144-3 2020 CYP2D6 metabolizes the conversion of MAP to amphetamine (AMP), while CYP2B6 and CYP3A4 predominantly mediate the conversion of DMS to AMP. Amphetamine 134-137 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 33008144-7 2020 DMS exposure increased due to a reduction in the abundance of CYP2B6 and CYP3A4 in RI and HI subjects. desmethylselegiline 0-3 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 62-68 32960272-0 2021 CYP2B6 Genotype and Weight Gain Differences Between Dolutegravir and Efavirenz. dolutegravir 52-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32960272-0 2021 CYP2B6 Genotype and Weight Gain Differences Between Dolutegravir and Efavirenz. efavirenz 69-78 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32960272-2 2021 Loss-of-function polymorphisms in CYP2B6 result in higher efavirenz concentrations, which we hypothesized would impair weight gain among people living with human immunodeficiency virus (HIV; PLWH) starting efavirenz-based antiretroviral therapy (ART). efavirenz 58-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 32960272-2 2021 Loss-of-function polymorphisms in CYP2B6 result in higher efavirenz concentrations, which we hypothesized would impair weight gain among people living with human immunodeficiency virus (HIV; PLWH) starting efavirenz-based antiretroviral therapy (ART). efavirenz 206-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 32960272-11 2021 CONCLUSIONS: CYP2B6 metaboliser genotype was associated with weight gain in PLWH starting efavirenz-based ART. efavirenz 90-99 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 32817462-0 2020 Tyrosine Nitration Contributes to Nitric Oxide-Stimulated Degradation of CYP2B6. Tyrosine 0-8 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 32817462-0 2020 Tyrosine Nitration Contributes to Nitric Oxide-Stimulated Degradation of CYP2B6. Nitric Oxide 34-46 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 32817462-1 2020 Human cytochrome P450 (P450) CYP2B6 undergoes nitric oxide (NO)-dependent proteasomal degradation in response to the NO donor dipropylenetriamine NONOate (DPTA) and biologic NO in HeLa and HuH7 cell lines. Nitric Oxide 46-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 32817462-9 2020 We propose that cumulative nitrations of Y190, Y317, and Y380 by reactive nitrogen species cause destabilization of CYP2B6, which may act synergistically with heme nitrosylation to target the enzyme for degradation. Nitrogen 74-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 32817462-1 2020 Human cytochrome P450 (P450) CYP2B6 undergoes nitric oxide (NO)-dependent proteasomal degradation in response to the NO donor dipropylenetriamine NONOate (DPTA) and biologic NO in HeLa and HuH7 cell lines. dipropylenetriamine-NONOate 126-153 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 32817462-9 2020 We propose that cumulative nitrations of Y190, Y317, and Y380 by reactive nitrogen species cause destabilization of CYP2B6, which may act synergistically with heme nitrosylation to target the enzyme for degradation. Heme 159-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 116-122 32817462-10 2020 SIGNIFICANCE STATEMENT: This work provides novel insight into the mechanisms by which nitric oxide, which is produced in hepatocytes in response to inflammation, triggers the ubiquitin-dependent proteasomal degradation of the cytochrome P450 (P450) enzyme CYP2B6. Nitric Oxide 86-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 256-262 32817462-1 2020 Human cytochrome P450 (P450) CYP2B6 undergoes nitric oxide (NO)-dependent proteasomal degradation in response to the NO donor dipropylenetriamine NONOate (DPTA) and biologic NO in HeLa and HuH7 cell lines. dipropylenetriamine-NONOate 155-159 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 32817462-3 2020 We hypothesized that NO or derivative reactive nitrogen species may generate adducts of tyrosine and/or cysteine residues, causing CYP2B6 downregulation, and selected Tyr and Cys residues for mutation based on predicted solvent accessibility. Nitrogen 47-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 32817462-6 2020 Carbon monoxide-releasing molecule 2 treatment caused rapid suppression of 2B6 enzyme activity, significant HMM species generation, and ubiquitination of CYP2B6 protein but did not stimulate CYP2B6 degradation. Carbon Monoxide 0-15 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 154-160 32817462-7 2020 The CYP2B6 inhibitor 4-(4-chlorophenyl)imidazole blocked NO-dependent CYP2B6 degradation, suggesting that NO access to the active site is important. 4-(4-chlorophenyl)imidazole 21-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 4-10 32817462-7 2020 The CYP2B6 inhibitor 4-(4-chlorophenyl)imidazole blocked NO-dependent CYP2B6 degradation, suggesting that NO access to the active site is important. 4-(4-chlorophenyl)imidazole 21-48 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 70-76 32817462-8 2020 Molecular dynamics simulations predicted that tyrosine nitrations of CYP2B6 would cause significant destabilizing perturbations of secondary structure and remove correlated motions likely required for enzyme function. Tyrosine 46-54 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 32903464-0 2020 Analysis of Drug Metabolizing Gene Panel in Osteosarcoma Patients Identifies Association Between Variants in SULT1E1, CYP2B6 and CYP4F8 and Methotrexate Levels and Toxicities. Methotrexate 140-152 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 118-124 32129697-5 2020 Rucaparib reversibly inhibited CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3As (IC50, 3.55, 12.9, 5.42, 41.6, and 17.2-22.9 microM [2 substrates], respectively), but not CYP2B6 or CYP2C8 (>190 microM). rucaparib 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 166-172 32129697-7 2020 In cultured human hepatocytes, rucaparib showed concentration-dependent induction of CYP1A2 mRNA and downregulation of CYP3A4 and CYP2B6 mRNA. rucaparib 31-40 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 130-136 32829410-5 2021 Associations were characterized with CYP2B6 and UGT1A1 genotypes that affect efavirenz and INSTI metabolism, respectively. efavirenz 77-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-43 32829410-10 2021 In the clinical trials cohort (N=462), CYP2B6 slow metabolizers had lesser weight gain at week 48 among participants receiving efavirenz with tenofovir disoproxil fumarate (p=0.001) but not those receiving efavirenz with abacavir (p=0.65). efavirenz 127-136 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 32829410-10 2021 In the clinical trials cohort (N=462), CYP2B6 slow metabolizers had lesser weight gain at week 48 among participants receiving efavirenz with tenofovir disoproxil fumarate (p=0.001) but not those receiving efavirenz with abacavir (p=0.65). Tenofovir 142-171 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 32829410-10 2021 In the clinical trials cohort (N=462), CYP2B6 slow metabolizers had lesser weight gain at week 48 among participants receiving efavirenz with tenofovir disoproxil fumarate (p=0.001) but not those receiving efavirenz with abacavir (p=0.65). efavirenz 206-215 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 32829410-10 2021 In the clinical trials cohort (N=462), CYP2B6 slow metabolizers had lesser weight gain at week 48 among participants receiving efavirenz with tenofovir disoproxil fumarate (p=0.001) but not those receiving efavirenz with abacavir (p=0.65). abacavir 221-229 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 39-45 32829410-12 2021 CONCLUSIONS: Among patients who switched from efavirenz- to INSTI-based therapy, CYP2B6 genotype was associated with weight gain, possibly reflecting withdrawal of the inhibitory effect of higher efavirenz concentrations on weight gain. efavirenz 46-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 32829410-12 2021 CONCLUSIONS: Among patients who switched from efavirenz- to INSTI-based therapy, CYP2B6 genotype was associated with weight gain, possibly reflecting withdrawal of the inhibitory effect of higher efavirenz concentrations on weight gain. efavirenz 196-205 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 81-87 32903464-12 2020 This is the first study to identify genetic variants in SULT1E1, CYP2B6, and CYP4F8 to be associated with methotrexate pharmacokinetics and toxicities. Methotrexate 106-118 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 65-71 32482757-0 2020 NADPH-Independent Inactivation of CYP2B6 and NADPH-dependent Inactivation of CYP3A4/5 by Pyrrolobenzodiazepine Dimer (PBD) and Potential Implication for Assessing Covalent Modulators for Time-dependent Inhibition. pyrrolo(2,1-c)(1,4)benzodiazepine 89-110 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 32377759-0 2020 Prediction of the exposure to a 400-mg daily dose of efavirenz in pregnancy: is this dose adequate in extensive metabolisers of CYP2B6? efavirenz 53-62 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 128-134 32564328-5 2020 Understanding of the mechanisms of variability in methadone disposition and drug interactions has evolved over the years, with the latest evidence revealing that CYP 2B6 is the major determinant of methadone elimination and plays a key role in methadone-related drug interactions. Methadone 50-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-169 32564328-5 2020 Understanding of the mechanisms of variability in methadone disposition and drug interactions has evolved over the years, with the latest evidence revealing that CYP 2B6 is the major determinant of methadone elimination and plays a key role in methadone-related drug interactions. Methadone 198-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-169 32564328-5 2020 Understanding of the mechanisms of variability in methadone disposition and drug interactions has evolved over the years, with the latest evidence revealing that CYP 2B6 is the major determinant of methadone elimination and plays a key role in methadone-related drug interactions. Methadone 198-207 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-169 32482757-4 2020 All other CYP isoforms tested did not show evidence for TDI, but potent inhibition of CYP2B6 (IC50 = 1.5 microM) was observed following a 30-minute pre-incubation both in the absence and presence of NADPH, an unexpected observation given the fact that no CYP2B6 inhibition was observed in the direct reversible inhibition assay up to 10 microM of PBD. 1,1'-((propane-1,3-diyl)dioxy)bis(7-methoxy-2-methylidene-1,2,3,10,11,11a-hexahydro-5H-pyrrolo(2,1-c)(1,4)benzodiazepin-5,11-dione) 347-350 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 86-92 32482757-5 2020 No other CYP isoforms were susceptible to this apparent non-NADPH dependent inhibition, suggesting that PBD may selectively inactivate CYP2B6 without metabolic activation. 1,1'-((propane-1,3-diyl)dioxy)bis(7-methoxy-2-methylidene-1,2,3,10,11,11a-hexahydro-5H-pyrrolo(2,1-c)(1,4)benzodiazepin-5,11-dione) 104-107 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 135-141 32482757-6 2020 The washing of the HLM pellet after 45 min incubation with PBD did not fully recover the CYP2B6 activity, indicating that PBD is covalently binding to CYP2B6 leading to inactivation of the enzyme. 1,1'-((propane-1,3-diyl)dioxy)bis(7-methoxy-2-methylidene-1,2,3,10,11,11a-hexahydro-5H-pyrrolo(2,1-c)(1,4)benzodiazepin-5,11-dione) 122-125 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 151-157 32209837-6 2020 Linear regressions, Mann-Whitney U-test or Kruskal-Wallis tests were conducted to examine the association of lumefantrine plasma level with CYP2B6 c.516G>T, NR1I3 152c-1089T>C, CYP2B6 c.983T>C, CYP3A5*3 and CYP3A4*22. Lumefantrine 109-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 140-146 32974447-10 2020 Genotype analysis of cytochrome P450 enzyme (CYP) demonstrated CYP2B6*1/*2 polymorphism leading to impaired metabolism of prasugrel. Prasugrel Hydrochloride 122-131 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 63-69 32415468-9 2020 CYP2B6 516GT carriers showed a significant reduction of imatinib concentration at 24 h (23%, 391 ng/dL vs 511 ng/dL in 516GG carriers, p = 0.005) and elimination half-life (11%, 12.6 h vs 14.1 h in 516GG carriers, p = 0.041). Imatinib Mesylate 56-64 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32415468-12 2020 CONCLUSION: CYP2B6 G516T and CYP3A4 *20,*22 polymorphisms could influence imatinib plasma concentrations and safety profile, after single-dose administration to healthy subjects. Imatinib Mesylate 74-82 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 32538291-0 2020 Involvement of CYP2D6 and CYP2B6 on tramadol pharmacokinetics. Tramadol 36-44 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 26-32 32538291-4 2020 CYP2B6 G516T T/T (n = 2) genotype was also associated to higher tramadol plasma levels. Tramadol 64-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32538291-7 2020 The correlation of CYP2B6 genotype with higher tramadol concentrations is remarkable since its influence on its elimination is also relevant and has been less studied to date. Tramadol 47-55 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 19-25 32838647-5 2020 CYP2B6 516G>T, 785A>G, 18492C>T and ABCB1 3435C>T T/C were associated with higher efavirenz plasma levels in the standard but not the lower-dose group. efavirenz 82-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32451120-2 2020 Polymorphism of CYP2B6 516G > T has been found to predominantly contribute to efavirenz variability. efavirenz 78-87 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 32451120-4 2020 This study aimed to develop a population model of the pharmacokinetic properties of efavirenz, and to investigate the impact of patients" characteristics and CYP2B6 516G > T polymorphism on the pharmacokinetic properties of efavirenz. efavirenz 224-233 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 158-164 32451120-9 2020 The efavirenz oral clearance were 11.9, 8.0, and 2.8 L/h in patients weighing 57 kg and having the CYP2B6 516 GG, 516 GT, and 516 TT genotypes, respectively. efavirenz 4-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 32451120-12 2020 IMPLICATION: The results from this study provide a rationale for efavirenz dose adjustment based on CYP2B6 516G > T polymorphism in Thai HIV-infected patients, which could help to improve treatment outcomes in this population. efavirenz 65-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-106 32209837-10 2020 CYP2B6 c.983T>C significantly associated with higher log Cday 7 of desbutyl lumefantrine in both pregnant (beta = 0.383; P = 0.033) and nonpregnant (beta = 0.395; P = 0.023) groups. desbutyllumefantrine 67-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32209837-11 2020 Composite genotypes for both CYP2B6 Single-nucleotide polymorphisms strongly associated with lumefantrine plasma concentration. Lumefantrine 93-105 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 29-35 32209837-13 2020 CONCLUSION: The findings revealed that the efavirenz-lumefantrine interaction was accentuated in the group with CYP2B6 c.516T, c.983C and NR1I3 152c-1089T alleles. efavirenz-lumefantrine 43-65 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 112-118 32209837-8 2020 In nonpregnant group, CYP2B6 c.516G>T was significantly associated with lower log Cday 7 of lumefantrine using multivariate linear regressions (beta = -0.239; P = 0.013). Lumefantrine 92-104 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 32209837-9 2020 In 59% of women with CYP2B6 c.516T, Cday 7 of lumefantrine was below the target of 280 ng/mL compared to 47% in the noncarriers. Lumefantrine 46-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 32364297-3 2020 Deoxyshikonin strongly inhibited CYP2B6-catalyzed bupropion hydroxylation, with a Ki value of 3.5 muM, and the inhibition was confirmed using purified human CYP2B6. deoxyshikonin 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 32234672-6 2020 IL-22 was next found to activate the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) 3 pathway, whose inhibition by the JAK inhibitor ruxolitinib fully prevented the IL-22-mediated CYP3A4, CYP2B6 and NTCP repression in HepaRG cells. ruxolitinib 161-172 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 216-222 32302325-8 2020 We assessed hepatic fibrosis and steatosis by transient elastography and CYP2B6 alleles, principally responsible for methadone metabolism. Methadone 117-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 73-79 32302325-13 2020 CYP2B6 loss of function (LOF) alleles significantly affected (S)-methadone metabolism (p = 0.012). Methadone 65-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32302325-17 2020 Although the principal methadone metabolizing enzyme remains controversial, our results suggest that sex, CYP2B6 genotype, and BMI should be incorporated into multivariate models to create methadone dosing algorithms. Methadone 189-198 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 106-112 32279544-3 2020 The aim of this study was to determine whether the presence of certain allelic variants in genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in Mexican children with solid tumours treated with ifosfamide.Objective: The aim of this study was to determine whether the presence of certain allelic variants of genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in children with solid tumours treated with ifosfamide.Materials and methods: A total of 131 DNA samples were genotyped by real-time polymerase chain reaction (RT-PCR) using TaqMan probes. Ifosfamide 214-224 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 32279544-3 2020 The aim of this study was to determine whether the presence of certain allelic variants in genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in Mexican children with solid tumours treated with ifosfamide.Objective: The aim of this study was to determine whether the presence of certain allelic variants of genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in children with solid tumours treated with ifosfamide.Materials and methods: A total of 131 DNA samples were genotyped by real-time polymerase chain reaction (RT-PCR) using TaqMan probes. Ifosfamide 214-224 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 333-339 32279544-3 2020 The aim of this study was to determine whether the presence of certain allelic variants in genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in Mexican children with solid tumours treated with ifosfamide.Objective: The aim of this study was to determine whether the presence of certain allelic variants of genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in children with solid tumours treated with ifosfamide.Materials and methods: A total of 131 DNA samples were genotyped by real-time polymerase chain reaction (RT-PCR) using TaqMan probes. Ifosfamide 442-452 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 97-103 32238417-0 2020 Stereoselective bupropion hydroxylation by cytochrome P450 CYP2B6 and cytochrome P450 oxidoreductase genetic variants. Bupropion 16-25 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 59-65 32238417-1 2020 Bioactivation of the antidepressant and smoking cessation drug bupropion is catalyzed predominantly by cytochrome P4502B6 (CYP2B6). Bupropion 63-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 103-121 32238417-1 2020 Bioactivation of the antidepressant and smoking cessation drug bupropion is catalyzed predominantly by cytochrome P4502B6 (CYP2B6). Bupropion 63-72 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 123-129 32238417-3 2020 Bupropion hydroxylation is the canonical in vitro and in vivo probe for CYP2B6 activity. Bupropion 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 72-78 32238417-6 2020 Some CYP2B6 variants affect bupropion disposition. Bupropion 28-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 5-11 32238417-7 2020 This investigation evaluated the influence of several human CYP2B6 and POR genetic variants on stereoselective bupropion metabolism, using an insect cell coexpression system containing CYP2B6, POR and cytochrome b5. Bupropion 111-120 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 60-66 32238417-10 2020 CYP2B6-catalyzed bupropion hydroxylation is stereoselective. Bupropion 17-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 32238417-12 2020 Established concordance between human bupropion hydroxylation in vitro and in vivo, together with these new results, suggests additional CYP2B6 variants may influence human bupropion disposition. Bupropion 38-47 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 32238417-12 2020 Established concordance between human bupropion hydroxylation in vitro and in vivo, together with these new results, suggests additional CYP2B6 variants may influence human bupropion disposition. Bupropion 173-182 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 137-143 32238417-13 2020 SIGNIFICANCE STATEMENT: Bupropion pharmacokinetics, metabolism and clinical effects are affected by the CYP2B6*6 polymorphism. Bupropion 24-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 104-110 32238417-14 2020 Other expressed CYP2B6 polymorphisms had diminished (*5, *6, *7, *9, *19, *26) or defective (*16, *18) in vitro bupropion hydroxylation. Bupropion 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 16-22 32238417-16 2020 These CYP2B6 polymorphisms may portend diminished in vivo bupropion hydroxylation and predict additional clinically important variant alleles. Bupropion 58-67 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 6-12 31772310-6 2020 We demonstrate that IV ketamine can be transitioned to oral regimen to shorten length of stay in the intensive care unit and hospital and has future CYP2B6 pharmacogenomic considerations for further dose individualization. Ketamine 23-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 32453717-11 2020 The clearance rate of EA in CYP2B6-expressing (p<0.05) and CYP2C9-expressing (p<0.001) HIEC cell models was remarkably reduced after 120 min. Ellagic Acid 22-24 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 32364297-3 2020 Deoxyshikonin strongly inhibited CYP2B6-catalyzed bupropion hydroxylation, with a Ki value of 3.5 muM, and the inhibition was confirmed using purified human CYP2B6. deoxyshikonin 0-13 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 32364297-3 2020 Deoxyshikonin strongly inhibited CYP2B6-catalyzed bupropion hydroxylation, with a Ki value of 3.5 muM, and the inhibition was confirmed using purified human CYP2B6. Bupropion 50-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 33-39 32364297-3 2020 Deoxyshikonin strongly inhibited CYP2B6-catalyzed bupropion hydroxylation, with a Ki value of 3.5 muM, and the inhibition was confirmed using purified human CYP2B6. Bupropion 50-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 157-163 32106141-8 2020 At day 21 in the efavirenz group, CYP2B6 genotype was associated with increased plasma efavirenz exposure (P = 3.2 x 10), decreased plasma concentrations of etonogestrel (P = 1.7 x 10), and decreased ethinyl estradiol (P = 6.7 x 10). efavirenz 17-26 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 30778771-0 2020 Association of SLC28A3 Gene Expression and CYP2B6*6 Allele with the Response to Fludarabine Plus Cyclophosphamide in Chronic Lymphocytic Leukemia Patients. fludarabine 80-91 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 30778771-0 2020 Association of SLC28A3 Gene Expression and CYP2B6*6 Allele with the Response to Fludarabine Plus Cyclophosphamide in Chronic Lymphocytic Leukemia Patients. Cyclophosphamide 97-113 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 43-49 31628422-0 2020 Influence of CYP2B6 activity score on the pharmacokinetics and safety of single dose efavirenz in healthy volunteers. efavirenz 85-94 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 13-19 31628422-2 2020 Cytochrome P450 (CYP) CYP2B6 G516T (rs3745274) is a well-known predictor of efavirenz disposition. efavirenz 76-85 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 22-28 31628422-5 2020 In this study, we evaluated the influence of 11 single-nucleotide polymorphisms (SNPs) in CYP2B6, CYP2A6, CYP3A and ABCB1 (ATP-binding cassette sub-family B member 1) on the pharmacokinetics and safety of efavirenz after single oral dose administration to 47 healthy volunteers. efavirenz 205-214 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 90-96 31628422-6 2020 We designed and validated a CYP2B6 activity score model based on two CYP2B6 SNPs (G516T and rs4803419) that predicted efavirenz disposition better than G516T alone. efavirenz 118-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 31628422-6 2020 We designed and validated a CYP2B6 activity score model based on two CYP2B6 SNPs (G516T and rs4803419) that predicted efavirenz disposition better than G516T alone. efavirenz 118-127 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 69-75 32106141-8 2020 At day 21 in the efavirenz group, CYP2B6 genotype was associated with increased plasma efavirenz exposure (P = 3.2 x 10), decreased plasma concentrations of etonogestrel (P = 1.7 x 10), and decreased ethinyl estradiol (P = 6.7 x 10). efavirenz 87-96 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 32106141-8 2020 At day 21 in the efavirenz group, CYP2B6 genotype was associated with increased plasma efavirenz exposure (P = 3.2 x 10), decreased plasma concentrations of etonogestrel (P = 1.7 x 10), and decreased ethinyl estradiol (P = 6.7 x 10). etonogestrel 157-169 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 32106141-8 2020 At day 21 in the efavirenz group, CYP2B6 genotype was associated with increased plasma efavirenz exposure (P = 3.2 x 10), decreased plasma concentrations of etonogestrel (P = 1.7 x 10), and decreased ethinyl estradiol (P = 6.7 x 10). Ethinyl Estradiol 200-217 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 34-40 32106141-9 2020 Compared to controls, efavirenz reduced median etonogestrel concentrations by at least 93% in CYP2B6 slow metabolizers versus approximately 75% in normal and intermediate metabolizers. efavirenz 22-31 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 32106141-9 2020 Compared to controls, efavirenz reduced median etonogestrel concentrations by at least 93% in CYP2B6 slow metabolizers versus approximately 75% in normal and intermediate metabolizers. etonogestrel 47-59 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 94-100 32106141-10 2020 Efavirenz reduced median ethinyl estradiol concentrations by 75% in CYP2B6 slow metabolizers versus approximately 41% in normal and intermediate metabolizers. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 32106141-10 2020 Efavirenz reduced median ethinyl estradiol concentrations by 75% in CYP2B6 slow metabolizers versus approximately 41% in normal and intermediate metabolizers. Ethinyl Estradiol 25-42 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 68-74 32106141-11 2020 CONCLUSION: CYP2B6 slow metabolizer genotype worsens the pharmacokinetic interaction of efavirenz with hormonal contraceptives administered by vaginal ring. efavirenz 88-97 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 32106141-12 2020 Efavirenz dose reduction in CYP2B6 slow metabolizers may reduce, but will likely not eliminate, this interaction. efavirenz 0-9 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 28-34 31967796-0 2020 Computational Biotransformation Profile of Emerging Phenolic Pollutants by Cytochromes P450: Phenol Coupling Mechanism. phenolic acid 52-60 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 31901712-0 2020 Retention of polybrominated diphenyl ethers and hydroxylated metabolites in paired human serum and milk in relation to CYP2B6 genotype. Phenyl Ethers 28-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 119-125 31901712-2 2020 Retention of PBDEs and OH-BDEs in humans may be affected by differences in PBDE metabolism due to variants in cytochrome P450 2B6 (CYP2B6). Halogenated Diphenyl Ethers 13-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-129 31901712-2 2020 Retention of PBDEs and OH-BDEs in humans may be affected by differences in PBDE metabolism due to variants in cytochrome P450 2B6 (CYP2B6). Halogenated Diphenyl Ethers 13-18 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 31901712-2 2020 Retention of PBDEs and OH-BDEs in humans may be affected by differences in PBDE metabolism due to variants in cytochrome P450 2B6 (CYP2B6). oh-bdes 23-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-129 31901712-2 2020 Retention of PBDEs and OH-BDEs in humans may be affected by differences in PBDE metabolism due to variants in cytochrome P450 2B6 (CYP2B6). oh-bdes 23-30 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 31901712-2 2020 Retention of PBDEs and OH-BDEs in humans may be affected by differences in PBDE metabolism due to variants in cytochrome P450 2B6 (CYP2B6). Halogenated Diphenyl Ethers 13-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 110-129 31901712-2 2020 Retention of PBDEs and OH-BDEs in humans may be affected by differences in PBDE metabolism due to variants in cytochrome P450 2B6 (CYP2B6). Halogenated Diphenyl Ethers 13-17 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 131-137 31901712-5 2020 Participants with CYP2B6*6 genotype had a greater relative retention of PBDEs in serum and milk, and significant relationships (p < 0.05) were also observed for PBDE-47, 5-OH-BDE-47 and 6-OH-BDE-47 concentrations relative to CYP2B6*5 and CYP2B6*6 genotypes. Halogenated Diphenyl Ethers 72-77 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 31901712-5 2020 Participants with CYP2B6*6 genotype had a greater relative retention of PBDEs in serum and milk, and significant relationships (p < 0.05) were also observed for PBDE-47, 5-OH-BDE-47 and 6-OH-BDE-47 concentrations relative to CYP2B6*5 and CYP2B6*6 genotypes. Halogenated Diphenyl Ethers 72-76 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 18-24 31901712-6 2020 These results are the first to show that CYP2B6 genotype is significantly related to the relative retention of PBDEs in humans, which may have direct implications for variability in the susceptibility of individuals to the potential adverse effects of these contaminants. Halogenated Diphenyl Ethers 111-116 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 41-47 31967796-0 2020 Computational Biotransformation Profile of Emerging Phenolic Pollutants by Cytochromes P450: Phenol Coupling Mechanism. Phenols 52-58 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 87-91 31967796-1 2020 Phenols are ubiquitous environmental pollutants, whose biotransformation involving phenol coupling catalyzed by cytochromes P450, may produce more lipophilic and toxic metabolites. Phenols 0-7 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 31967796-1 2020 Phenols are ubiquitous environmental pollutants, whose biotransformation involving phenol coupling catalyzed by cytochromes P450, may produce more lipophilic and toxic metabolites. Phenols 83-89 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 124-128 31967796-6 2020 Therefore, oxidation of triclosan by P450 fits the first equation with a ratio [coupling]/[hydroxylation] of 1:4, consistent with experimental data indicating different extents of triclosan coupling (6-40%). Triclosan 24-33 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-41 31967796-6 2020 Therefore, oxidation of triclosan by P450 fits the first equation with a ratio [coupling]/[hydroxylation] of 1:4, consistent with experimental data indicating different extents of triclosan coupling (6-40%). Triclosan 180-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 37-41 31924695-0 2020 Ser100-Phosphorylated RORalpha Orchestrates CAR and HNF4alpha to Form Active Chromatin Complex in Response to Phenobarbital to Regulate Induction of CYP2B6. ZP120 0-6 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 31924695-2 2020 Here, we found serine 100-phosphorylated RORalpha orchestrates constitutive androstane receptor (CAR) and hepatocyte nuclear factor 4 alpha (HNF4alpha) to induce CYP2B6 by phenobarbital (PB) in human primary hepatocytes (HPHs). Serine 15-21 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 32131538-5 2020 These in vitro DDI potentials of mertansine with CYP1A2, CYP2B6, CYP2C8/9/19, CYP3A4, UGT1A1, and UGT1A9 substrates suggest that it is necessary to carefully characterize the DDI potentials of ADC candidates with mertansine as a payload in the clinic. Maytansine 33-43 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 57-63 31549442-1 2020 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of the antimalarial drugs artemether and lumefantrine. artemether 85-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 31549442-1 2020 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of the antimalarial drugs artemether and lumefantrine. artemether 85-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 31549442-1 2020 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of the antimalarial drugs artemether and lumefantrine. lumefantrine 100-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-19 31549442-1 2020 Cytochrome P450 2B6 (CYP2B6) is involved in the metabolism of the antimalarial drugs artemether and lumefantrine. lumefantrine 100-112 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 21-27 31549442-2 2020 Here we investigated the effect of CYP2B6*6 on the plasma pharmacokinetics of artemether, lumefantrine, and their metabolites in healthy volunteers. artemether 78-88 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 35-41 31549442-12 2020 This is the first study in humans to associate CYP2B6*6 genotype with artemether disposition. artemether 70-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 47-53 31924695-2 2020 Here, we found serine 100-phosphorylated RORalpha orchestrates constitutive androstane receptor (CAR) and hepatocyte nuclear factor 4 alpha (HNF4alpha) to induce CYP2B6 by phenobarbital (PB) in human primary hepatocytes (HPHs). Phenobarbital 172-185 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 31924695-0 2020 Ser100-Phosphorylated RORalpha Orchestrates CAR and HNF4alpha to Form Active Chromatin Complex in Response to Phenobarbital to Regulate Induction of CYP2B6. Phenobarbital 110-123 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 149-155 31924695-2 2020 Here, we found serine 100-phosphorylated RORalpha orchestrates constitutive androstane receptor (CAR) and hepatocyte nuclear factor 4 alpha (HNF4alpha) to induce CYP2B6 by phenobarbital (PB) in human primary hepatocytes (HPHs). Phenobarbital 187-189 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 162-168 30902024-14 2020 These findings demonstrate that the relative CYP3A4, CYP2B6, and CYP2C19 involvement in meperidine N-demethylation depends on the enzyme activities in individual human liver microsomal samples. Meperidine 88-98 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 31924695-9 2020 Altogether, the results established that p-Ser100 RORalpha bridging the PBREM and OARE orchestrates CAR and HNF4alpha to form active chromatin complex during PB-induced CYP2B6 expression in human primary hepatocytes. p-ser100 41-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 31924695-9 2020 Altogether, the results established that p-Ser100 RORalpha bridging the PBREM and OARE orchestrates CAR and HNF4alpha to form active chromatin complex during PB-induced CYP2B6 expression in human primary hepatocytes. Phenobarbital 72-74 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 169-175 31924695-10 2020 SIGNIFICANCE STATEMENT: CYP2B6 is a vital enzyme for the metabolic elimination of xenobiotics, and it is prone to induction by xenobiotics, including phenobarbital via constitutive androstane receptor (CAR) and hepatocyte nuclear factor 4 alpha (HNF4alpha). Phenobarbital 150-163 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 24-30 32033200-10 2020 Combined, a higher rate of patients with CYP2W1*6WT+CYP2B6*6 (60.6%) achieved mitotane therapeutic range (p = 0.034). Mitotane 78-86 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 52-58 30902024-3 2020 Previous studies indicate that meperidine N-demethylation is catalyzed by cytochrome P450 2B6 (CYP2B6), CYP3A4, and CYP2C19. Meperidine 31-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 74-93 30902024-3 2020 Previous studies indicate that meperidine N-demethylation is catalyzed by cytochrome P450 2B6 (CYP2B6), CYP3A4, and CYP2C19. Meperidine 31-41 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 95-101 32433020-11 2020 Distinct expression of UGT1A1, CYP2B6, CYP2C9, CYP3A4, and CYP7A1 genes explains the ability to clear toxins and bilirubin as observed in normal hepatocytes. Bilirubin 113-122 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 31-37 30902024-8 2020 The catalytic efficiency (kcat/Km) for meperidine N-demethylation was similar between recombinant CYP2B6 and CYP2C19, but markedly lower by CYP3A4. Meperidine 39-49 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 30902024-14 2020 These findings demonstrate that the relative CYP3A4, CYP2B6, and CYP2C19 involvement in meperidine N-demethylation depends on the enzyme activities in individual human liver microsomal samples. Nitrogen 99-100 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 53-59 30902024-8 2020 The catalytic efficiency (kcat/Km) for meperidine N-demethylation was similar between recombinant CYP2B6 and CYP2C19, but markedly lower by CYP3A4. Nitrogen 50-51 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 98-104 32021384-1 2020 Purpose: This study was conducted to determine the effect of UGT1A9 98T>C, CYP2B6 516G>T and CYP2C9 430C>T genetic polymorphisms on the pharmacokinetics of propofol in children of different sexes and ages who undergone total intravenous anesthesia (TIVA) and deep sedation during diagnostic and therapeutic procedures. Propofol 156-164 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 75-81 31610395-8 2020 The abundance of CYP2B6 was 7.4 +- 7.8 pmol mg-1 microsomal protein and showed good correlation with activity against mephenytoin (Rs = 0.91, p < 0.0001; R2 = 0.93). Mephenytoin 118-129 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 17-23 32021384-7 2020 The carriers of the polymorphic CYP2B6 T allele received a significantly lower overall and initial dose of propofol. Propofol 107-115 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 32-38 32021384-9 2020 Conclusion: Further investigations of UGT1A9, CYP2B6 and CYP2C9 and other genes that participate in propofol metabolism as well as detailed analyses of the general conditions, administered therapies and associated diseases could explain the large interindividual variability of propofol metabolism in children. Propofol 100-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31706844-5 2020 By treatment with 3-deazaadenosine, an inhibitor of RNA methylation, CYP1A2, CYP2B6, and CYP2C8 levels were significantly increased (1.6-fold, 2.2-fold, and 2.7-fold, respectively) in HepaRG cells. 3-deazaadenosine 18-34 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 77-83 31633365-6 2019 In incubation experiments with eight recombinant CYP450s, we found that brivanib potently inhibited CYP2C subfamily members and the CYP2B6 isoform. brivanib 72-80 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 132-138 31402421-6 2019 RESULTS: Patients with AG or GG genotypes for CYP2B6 rs8109525 had a higher success rate of smoking cessation with varenicline (51.2%) compared with carriers of the AA genotypes (33.3%, P = 0.03, n = 167). Varenicline 115-126 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 46-52 31402421-8 2019 CONCLUSION: CYP2B6 rs8109525 was associated with a higher success rate of smoking cessation with varenicline treatment. Varenicline 97-108 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 12-18 31686598-0 2019 Severe and prolonged cyclophosphamide-induced hepatotoxicity in a breast cancer patient carrying a CYP2B6*7 variant. Cyclophosphamide 21-37 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 99-105 31686598-7 2019 CYP2B6 and ALDH3A1 genotyping may play a role in guiding cyclophosphamide therapy. Cyclophosphamide 57-73 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 0-6 30489204-13 2019 In addition, recombinant enzyme incubation experiments demonstrated that CYP3A4 was the predominant P450 responsible for the metabolism of silymarin. Silymarin 139-148 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 100-104 30489204-15 2019 Several P450 enzymes were reportedly inactivated by some of bioactive constituents of silymarin to some extent. Silymarin 86-95 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 8-12 30489204-16 2019 Our findings facilitate the understanding of mechanisms of the reported inactivation of P450 enzymes induced by silymarin. Silymarin 112-121 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 88-92