Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem J ; 355(Pt 2): 537-44, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11284743

RESUMO

Human dihydrodiol dehydrogenase (DD) 4/aldo-keto reductase (AKR) 1C4 is a major isoform of hepatic DD that oxidizes trans-dihydrodiols of polycyclic aromatic hydrocarbons to reactive and redox-active o-quinones and that reduces several ketone-containing drugs. To investigate the mechanism of transcriptional regulation of the human DD4 gene, the 5'-flanking region of the gene was fused to the luciferase gene. The results of luciferase assays using HepG2 cells and of 1,10-phenanthroline-copper footprinting indicated that two positive regulatory regions were located in regions from -701 to -684 and from -682 to -666. The former region contained a putative hepatocyte nuclear factor (HNF)-4 binding motif, and the latter region contained an HNF-1 consensus binding sequence. DNA fragments of the HNF-4 or HNF-1 motif gave a shifted band in a gel-shift assay with nuclear extracts from HepG2 cells. The formation of the DNA-protein complex was inhibited by the HNF-4 or HNF-1 motif of the alpha(1)-antitrypsin gene. A supershift assay using antibodies to human HNF-4alpha, HNF-4gamma and HNF-1alpha showed that HNF-4alpha and HNF-4gamma bound to the HNF-4 motif, and that HNF-1alpha interacted with the HNF-1 motif. Introduction of mutations into the HNF-4 or HNF-1 motif lowered the luciferase activity to 10 or 8% respectively of that seen with the intact human DD4 gene. These results indicate that HNF-4alpha, HNF-4gamma and HNF-1alpha regulate co-operatively the transcription of the human DD4 gene in HepG2 cells.


Assuntos
Oxirredutases do Álcool/genética , Proteínas de Ligação a DNA , Regulação Enzimológica da Expressão Gênica/fisiologia , Proteínas Nucleares , Oxirredutases/genética , Fosfoproteínas/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Aldeído Redutase , Aldo-Ceto Redutases , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Linhagem Celular , DNA , Pegada de DNA , Fator 1 Nuclear de Hepatócito , Fator 1-alfa Nuclear de Hepatócito , Fator 1-beta Nuclear de Hepatócito , Fator 4 Nuclear de Hepatócito , Humanos , Dados de Sequência Molecular , Mutação , Fenantrolinas/química , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Deleção de Sequência , Fatores de Transcrição/metabolismo
2.
Comp Biochem Physiol C Toxicol Pharmacol ; 126(3): 225-34, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11048672

RESUMO

Expression of drug-metabolizing enzymes including cytochrome P450 (CYP) and flavin-containing monooxygenase (FMO) in various tissues of Suncus murinus (Suncus) were examined. Northern blot analysis showed that mRNAs hybridizable with cDNAs for rat CYP1A2, human CYP2A6, rat CYP2B1, human CYP2C8, human CYP2D6, rat CYP2E1, human CYP3A4 and rat CYP4A1 were expressed in various tissues from Suncus. The mRNA level of CYP2A in the Suncus lung was very high. Furthermore, it was found that the level of CYP2A mRNA in the Suncus lung was higher compared to the Suncus liver. The expression level of mRNA hybridizable with cDNA for human CYP3A4 was very low. The presence of CYP3A gene in Suncus was proven by the induction of the CYP with dexamethasone. Very low expression levels of mRNAs hybridizable with cDNAs for rat FMO1, rat FMO2, rat FMO3 and rat FMO5 were also seen in Suncus liver. No apparent hybridization band appeared when human FMO4 cDNA was used as a probe. The hepatic expression of mRNAs hybridizable with cDNAs for UDP-glucuronosyltransferase 1*6, aryl sulfotransferase, glutathione S-transferase 1, carboxyesterase and microsomal epoxide hydrolase in the Suncus were observed. These results indicate that the Suncus is a unique animal species in that mRNAs for CYP3A and FMO are expressed at very low levels.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/genética , Eulipotyphla/genética , Expressão Gênica , Oxirredutases N-Desmetilantes/genética , Oxigenases/genética , Animais , Northern Blotting , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/metabolismo , Dexametasona/farmacologia , Eulipotyphla/metabolismo , Moduladores GABAérgicos/metabolismo , Expressão Gênica/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Midazolam/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Oxigenases/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo
3.
Mutat Res ; 428(1-2): 125-30, 1999 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-10517986

RESUMO

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). The 5'-terminus of CYP cDNA was modified to achieve a high-level expression in S. typhimurium. Modified CYP2A6 or CYP2E1 cDNA and native OR cDNA were introduced into a pCW vector. S. typhimurium YG7108 cells were transformed with this vector. The mutagen producing ability of these enzymes for some N-nitrosamines were evaluated using the established S. typhimurium cells. We found that the substrate specificity of CYP2A6 and CYP2E1 was different among mutagens. CYP2A6 was responsible for the metabolic activation of N-nitrosamines possessing relatively long alkyl chains, whereas CYP2E1 was responsible for the metabolic activation of N-nitrosamines with relatively short alkyl chains. It is likely that CYP2A6 gene polymorphism is responsible for the interindividual variability on the cancer susceptibility. We found the whole deletion of CYP2A6 gene as a type of genetic polymorphism in Japanese. Thus, we developed a gene diagnosis method to detect the variant. We evaluated the relationship between the CYP2A6 gene whole deletion and the susceptibility to the lung cancer. The frequency of CYP2A6 gene whole deletion was significantly lower in the lung cancer patients than that of healthy volunteers.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/genética , Oxigenases de Função Mista/genética , Neoplasias/enzimologia , Neoplasias/genética , Polimorfismo Genético , Estudos de Casos e Controles , Citocromo P-450 CYP2A6 , Citocromo P-450 CYP2E1/genética , Deleção de Genes , Expressão Gênica , Frequência do Gene , Humanos , Japão , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Testes de Mutagenicidade , NADPH-Ferri-Hemoproteína Redutase/genética , Salmonella typhimurium/genética
4.
J Biochem ; 126(2): 402-7, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10423536

RESUMO

To clarify the molecular mechanisms involved in the generation of the CYP2A6 gene deletion (E-type variant), we analyzed the CYP2A7 gene, which is located in the 5'-flanking region of the CYP2A6 gene, from individuals with the E-type variant and compared it with the sequences of wild type CYP2A7 and CYP2A6 genes. The 3'-downstream sequence (up to 324 bp from the SacI site in exon 9) of the CYP2A7 gene of the E-type variant is identical to that of the wild CYP2A7 gene. However, the 3'-downstream sequence (starting from 325 bp from the SacI site in exon 9) of the CYP2A7 gene of the E-type variant is identical to that of the wild CYP2A6 gene, indicating that the 3'-downstream region of CYP2A7 and the 3'-downstream region of CYP2A6 linked directly eliminating the whole CYP2A6 gene. PCR analysis using primers specific to the CYP2A7 gene and the CYP2A6 and CYP2A7 genes confirmed that all DNA samples obtained from 7 individuals carrying the E-type variant possessed the same break points. These results indicate that the breakpoint of the CYP2A6 gene deletion lies in the 3'-downstream region of the CYP2A7 and CYP2A6 genes.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Troca Genética , Sistema Enzimático do Citocromo P-450/genética , Deleção de Genes , Oxigenases de Função Mista/genética , Família Multigênica , Esteroide Hidroxilases/genética , Alelos , Sequência de Bases , Southern Blotting , Citocromo P-450 CYP2A6 , Família 2 do Citocromo P450 , Éxons , Biblioteca Gênica , Humanos , Linfócitos , Modelos Genéticos , Dados de Sequência Molecular , Fenótipo , Polimorfismo Genético , Polimorfismo de Fragmento de Restrição , Alinhamento de Sequência
5.
J Pharmacol Exp Ther ; 289(1): 437-42, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10087035

RESUMO

(+)-Cis-3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one hydrochloride (SM-12502) is a newly developed drug as a platelet-activating factor receptor antagonist. The disposition of SM-12502 was investigated in plasma from 28 healthy Japanese volunteers after a single i.v. administration of SM-12502. Three of 28 subjects were phenotyped as poor metabolizers (PMs). Genomic DNAs from three extensive metabolizers or three PMs of SM-12502 were analyzed by Southern blot analysis with CYP2A6 cDNA as a probe. DNAs from three PMs digested with SacI and SphI showed novel restriction fragment length polymorphisms (RFLPs); one type without 4.5- and 2.6-kb fragments and a weak density of a 6.4-kb fragment (E-type), and the other type without 7.1- and 5.5-kb restriction fragments (C'-type) as compared with three extensive metabolizers, respectively. The deletional restriction fragments specific to three PMs in SacI- and SphI-RFLPs were identified as CYP2A6. Using polymerase chain reaction-RFLP analyses of the gene from the three PMs, we found that the exon 1, exon 8, and exon 9 in CYP2A6 were absent. A new RFLP characterized by SacI and SphI was found to be due to the entire gene deletion of the three exons and was associated with the decreased metabolism of SM-12502. This study demonstrates a new deletional allele in the human CYP2A6 gene responsible for the poor metabolic phenotype of SM-12502.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/genética , Oxigenases de Função Mista/genética , Inibidores da Agregação Plaquetária/metabolismo , Polimorfismo de Fragmento de Restrição , Deleção de Sequência , Tiazóis/metabolismo , Adulto , Alelos , Sequência de Bases , Southern Blotting , Citocromo P-450 CYP2A6 , DNA/genética , DNA/isolamento & purificação , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Éxons , Biblioteca Gênica , Humanos , Leucócitos/metabolismo , Masculino , Dados de Sequência Molecular , Fenótipo , Inibidores da Agregação Plaquetária/sangue , Inibidores da Agregação Plaquetária/urina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tiazóis/sangue , Tiazóis/urina , Tiazolidinas
6.
Pharmacogenetics ; 8(3): 239-49, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9682269

RESUMO

The S-oxidation of (4)-cis-3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one hydrochloride (SM-12502) and the 7-hydroxylation of coumarin are primarily catalyzed by cytochrome P450 2A6 (CYP2A6). The activities of SM-12502 S-oxidase and coumarin 7-hydroxylase were investigated with liver microsomes from 20 human individuals. Liver microsomes from individual H16 showed the lowest activities of both enzymes. The expression of CYP2A6 protein was not detectable in liver microsomes from individuals H4, H5, H7, H8, H12 and H16. CYP2A6 mRNA was hardly detectable in the liver of the individual H16. A new SacI-restriction fragment length polymorphism showing the lack of a 2.6 kb fragment was found in two of forty genomic DNA preparations from individuals H16 and No. 594, using CYP2A6 cDNA as a probe. This deletional 2.6 kb fragment was isolated from a genomic library prepared from one individuals showing normal coumarin 7-hydroxylase activity and was sequenced. This fragment contained a CYP2A6 gene region from 319 bp upstream of a putative exon 6 to a SacI site in exon 9, indicating that this region was deleted in the two individuals in this study. We also demonstrated by polymerase chain reaction analysis that the exon 8 of CYP2A6 gene was deleted in individuals H16 and No. 594. These results indicate that the reduced activity of SM-12502 S-oxidase and no activity of coumarin 7-hydroxylase are caused by the lack of CYP2A6 mRNA and CYP2A6 protein caused by the CYP2A6 gene deletion.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Cumarínicos/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Deleção de Genes , Oxigenases de Função Mista/genética , Tiazóis/metabolismo , Sequência de Bases , Clonagem Molecular , Citocromo P-450 CYP2A6 , Sistema Enzimático do Citocromo P-450/análise , Biblioteca Genômica , Humanos , Hidroxilação , Microssomos Hepáticos/metabolismo , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo de Fragmento de Restrição , Análise de Sequência de DNA , Especificidade por Substrato , Tiazolidinas
7.
Drug Metab Dispos ; 24(11): 1212-7, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8937855

RESUMO

Nicotine is primarily metabolized to cotinine in humans. In this study, human cytochrome P450 (CYP) isoform involved in cotinine formation was identified. The formation of cotinine in 16 human liver microsomes was determined with a 50 microM nicotine concentration and with a cytosol preparation as a source of aldehyde oxidase. Cotinine formation in human liver microsomes significantly correlated with immunochemically determined CYP2A6 levels (r = 0.663, p < 0.05), coumarin 7-hydroxylase activities (r = 0.831, p < 0.01), and cotinine 3'-hydroxylase activities (r = 0.735, p < 0.01) that are responsible for CYP2A6. In inhibition studies, cotinine formation in human liver microsomes was inhibited by coumarin and rabbit anti-rat CYP2A1 antibody specifically. When the capability of microsomes of B-lymphoblastoid cells expressing human CYPs to perform biotransformation of nicotine to cotinine was determined, cDNA-expressed CYP2A6 exhibited the highest cotinine formation. The KMapp values from microsome expressing CYP2A6 cDNA were similar to the value obtained from human liver microsomes. The large interindividual variabilities in cotinine formation and immunochemically determined CYP2A6 levels were observed in human liver microsomes, suggesting genetic polymorphism of CYP2A6. Nicotine is a new in vivo probe for phenotyping of CYP2A6 in humans.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/metabolismo , Oxigenases de Função Mista/metabolismo , Nicotina/metabolismo , Linfócitos B/enzimologia , Linfócitos B/metabolismo , Cotinina/metabolismo , Citocromo P-450 CYP2A6 , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/farmacologia , Humanos , Hidroxilação , Imunoquímica , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Oxigenases de Função Mista/antagonistas & inibidores , Oxirredução
8.
J Pharmacol Exp Ther ; 277(2): 1010-5, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8627511

RESUMO

Nicotine is primarily metabolized to cotinine, and cotinine is further metabolized to trans-3'-hydroxycotinine in human liver, which is a major metabolite of nicotine in humans. We studied the formation of trans-3'-hydroxycotinine from cotinine in human liver microsomes. trans-3'-Hydroxycotinine formation demonstrated single enzyme Michaelis-Menten kinetics (Km, 234.5 +/- 26.8 MicroM; Vmax, 37.2 +/- 2.4 pmol/min/mg protein). Significant correlation (r = .967, P < .001) between cotinine 3'-hydroxylase activities at low (50 microM) and high (1 microM) cotinine concentrations in 20 human liver microsomes suggested the contribution of a single enzyme to cotinine 3'-hydroxylation. The cotinine 3'-hydroxylase activity correlated significantly with immunoreactive cytochrome P450 (CYP)2A6 contents (r = .756, P < .01) and coumarin 7-hydroxylase activity (r = .887, P < .001). The cotinine 3'-hydroxylase activity was inhibited by coumarin, alpha-naphthoflavone, chlorzoxazone and anti-rat CYP2A1 antibodies. Microsomes of B-lymphoblastoid cells expressing human CYP2A6 exhibited cotinine 3'-hydroxylase activity. The Km value of the expressed CYP2A6 (264.7 microM) was almost identical to that of human liver microsomes. In conclusion, cotinine 3'-hydroxylation appears to be catalyzed solely by CYP2A6 in humans. Cotinine is a candidate for a new substrate for CYP2A6 in humans.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Cotinina/metabolismo , Sistema Enzimático do Citocromo P-450/fisiologia , Microssomos Hepáticos/metabolismo , Oxigenases de Função Mista/fisiologia , Citocromo P-450 CYP2A6 , Inibidores das Enzimas do Citocromo P-450 , Humanos , Hidroxilação , Isoenzimas/fisiologia , Oxigenases de Função Mista/antagonistas & inibidores
9.
J Pharmacol Exp Ther ; 277(2): 768-74, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8627557

RESUMO

(+)-cis-3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one hydrochloride (SM-12502) was oxidized by human liver microsomes to produce the S-oxide as a sole metabolite. Indirect evidence suggested that the S-oxidation was catalyzed by cytochrome P450 (CYP). Eadie-Hofstee plots showed biphasic pattern, suggesting that at least two enzymes were involved in the S-oxidation in human liver microsomes. Kinetic parameters of the S-oxidase with high-affinity showed Km and Vmax values of 20.9 +/- 4.4 microM and 0.111 +/- 0.051 nmol/min/mg microsomal protein, respectively. The S-oxidase activity was inhibited by coumarin and anti-CYP2A antibody. Among the contents of forms of CYP 20 samples of human liver microsomes, the content of CYP2A6 correlated with S-oxidase activity measured with 50 microM SM-12502 (r = .808, P < .0005). A close correlation (r = .908, P < .0001) was observed between activities of SM-12502 S-oxidase and coumarin 7-hydroxylase. Microsomes from genetically engineered human B-lymphoblastoid cells expressing CYP2A6 metabolized SM-12502 to the S-oxide efficiently. The results indicate that CYP2A6 isozyme is a major form of CYP responsible for the S-oxidation of SM-12502 in human liver microsomes. Thus, SM-12502 will be a useful tool in further research to analyze a human genetic polymorphism of CYP2A6.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/fisiologia , Oxigenases de Função Mista/fisiologia , Tiazóis/metabolismo , Animais , Citocromo P-450 CYP2A6 , Humanos , Isoenzimas/análise , Microssomos Hepáticos/metabolismo , Oxirredução , Ratos , Tiazolidinas
10.
Xenobiotica ; 25(12): 1283-91, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8719904

RESUMO

1. Rat hepatic flavin-containing monooxygenase 1 (FMO1) expressed in yeast catalyzed the S-oxidation of (+)-cis-3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one hydrochloride (SM-12502) in vitro. 2. S-oxidation was inhibited by 1-(1-naphthyl)-2-thiourea and thiobenzamide, known inhibitors of FMO, but was not enhanced by n-octylamine, a known enhancer of FMO. 3. The rate of S-oxide formation from SM-12502 was about four-fold lower than that from (+/-)-trans-3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one hydrochloride (SM-9979) and enantioselectivity and diastereoselectivity of the S-oxidation reaction were observed. 4. The ability of the recombinant yeast to produce the S-oxide from SM-12502 was maintained for long periods and exemplifies the recombinant yeast as a bioreactor to produce a large amount of the S-oxide.


Assuntos
Microssomos Hepáticos/enzimologia , Oxigenases/metabolismo , Saccharomyces cerevisiae/enzimologia , Tiazóis/metabolismo , Animais , Concentração de Íons de Hidrogênio , Oxirredução , Oxigenases/genética , Fator de Ativação de Plaquetas/antagonistas & inibidores , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Tiazolidinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...