Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Drug Metab Pharmacokinet ; 32(1): 100-107, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28153492

ABSTRACT

Sex and age differences in hepatic expression of drug-metabolizing enzyme genes could cause variations in drug metabolism, but has not been fully elucidated, especially in Asian population. In this study, the global expression of human hepatic genes was analyzed by microarrays in 40 Japanese subjects (27 males and 13 females). Thirty-five sex-biased genes were identified (P < 0.005). Whereas, 60 age-biased genes in two age groups, <60 years and ≥70 years (P < 0.001), were identified in males. By Gene Ontology analysis, the sex-biased genes were related to protein catabolism and modification, while the age-biased genes were related to transcription regulation and cell death. Quantitative polymerase chain reaction confirmed the female-biased expression of drug-metabolizing enzyme genes BChE, CYP4X1, and SULT1E1 (≥1.5-fold, P < 0.05). Further analysis of drug-metabolizing enzyme genes indicated that expression of CYP2A6 and CYP3A4 in females in the ≥70 age group was less than in the <60 age group (≥1.5-fold, P < 0.05), and this trend was also observed for PXR expression in males (≥1.5-fold, P < 0.05). The results presented provide important insights into hepatic physiology and function, especially drug metabolism, with respect to sex and age.


Subject(s)
Aging/genetics , Butyrylcholinesterase/metabolism , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Enzymologic , Liver/metabolism , Pharmaceutical Preparations/metabolism , Receptors, Steroid/metabolism , Sulfotransferases/metabolism , Aged , Butyrylcholinesterase/genetics , Cytochrome P-450 Enzyme System/genetics , Female , Gene Expression Profiling , Humans , Liver/enzymology , Liver/pathology , Liver/surgery , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Pregnane X Receptor , Receptors, Steroid/genetics , Sex Characteristics , Sulfotransferases/genetics
2.
Sci Rep ; 6: 21419, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26903378

ABSTRACT

Thalidomide is a teratogen in humans but not in rodents. It causes multiple birth defects including malformations of limbs, ears, and other organs. However, the species-specific mechanism of thalidomide teratogenicity is not completely understood. Reproduction of the human teratogenicity of thalidomide in rodents has previously failed because of the lack of a model reflecting human drug metabolism. In addition, because the maternal metabolic effect cannot be eliminated, the migration of unchanged thalidomide to embryos is suppressed, and the metabolic activation is insufficient to develop teratogenicity. Previously, we generated transchromosomic mice containing a human cytochrome P450 (CYP) 3A cluster in which the endogenous mouse Cyp3a genes were deleted. Here, we determined whether human CYP3A or mouse Cyp3a enzyme expression was related to the species difference in a whole embryo culture system using humanized CYP3A mouse embryos. Thalidomide-treated embryos with the human CYP3A gene cluster showed limb abnormalities, and human CYP3A was expressed in the placenta, suggesting that human CYP3A in the placenta may contribute to the teratogenicity of thalidomide. These data suggest that the humanized CYP3A mouse is a useful model to predict embryonic toxicity in humans.


Subject(s)
Abnormalities, Drug-Induced/pathology , Cytochrome P-450 CYP3A/genetics , Embryo, Mammalian/drug effects , Placenta/drug effects , Teratogens/toxicity , Thalidomide/toxicity , Animals , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Extremities , Female , Gene Expression , Humans , Inactivation, Metabolic , Mice , Mice, Transgenic , Placenta/enzymology , Pregnancy , Species Specificity , Transgenes
3.
Drug Metab Lett ; 6(2): 67-72, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22849705

ABSTRACT

Phenotyping of cytochrome P450 2A6 (CYP2A6) was determined by assessing urinary caffeine metabolites in a Japanese population with a high frequency of CYP2A6 whole-gene deletion (CYP2A6*4). The levels of 1,7-dimethyluric acid (17U), 1-methylxanthine (1X), and 1,7-dimethylxanthine (17X) were measured in non-smokers whose CYP2A6 and NAT2 genotypes had been determined. Low 17U/1X ratios were observed in accumulated overnight urine samples of subjects genotyped as CYP2A6*4/*4 after caffeine treatment. The individual 17U/1X ratios in spot urine samples were almost constant before and 2-8 h after caffeine treatment, with or without prior abstention from dietary caffeine. The average 17U/1X ratios obtained from subjects with CYP2A6 *4/ *4 or CYP2A6 *1/ *4 genotypes were significantly lower than those from subjects with wild-type CYP2A6 *1/ *1 under dietary caffeine consumption. The present results suggest that impaired CYP2A6 function associated with CYP2A6 *4/ *4 could be determined using the 17U/1X ratios in spot urine samples under normal dietary caffeine consumption in Japanese non-smokers, without the need for additional caffeine administration or prior abstention from caffeine.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Caffeine/metabolism , Theophylline/urine , Uric Acid/analogs & derivatives , Xanthines/urine , Adult , Caffeine/administration & dosage , Cytochrome P-450 CYP2A6 , Female , Gene Deletion , Genotype , Humans , Japan , Male , Phenotype , Pilot Projects , Uric Acid/urine , Young Adult
4.
J Toxicol Sci ; 37(3): 555-63, 2012.
Article in English | MEDLINE | ID: mdl-22687995

ABSTRACT

We previously found that genetic polymorphism in cytochrome P450 2A6 (CYP2A6) is one of the potential determinants of tobacco-related lung cancer risk. It has been reported that the plasma concentration of cotinine, a major metabolite of nicotine, in carriers of wild-type alleles of CYP2A6 is considerably higher than that in carriers of null or reduced-function alleles of CYP2A6, raising the possibility that cotinine plays an important role in the development of lung cancer. As a novel mechanism of lung tumorigenesis mediated by CYP2A6, we investigated the effects of cotinine on the suppression of apoptosis and promotion of lung tumor growth. In human lung adenocarcinoma A549 cells, cotinine inhibited doxorubicin-induced cell death by suppressing caspase-mediated apoptosis. Enhanced phosphorylation of Akt, a key factor responsible for cell survival and inhibition of apoptosis, was detected after cotinine treatment. These data suggest that cotinine suppresses caspase-mediated apoptosis induced by doxorubicin through activation of the PI3K/Akt pathway. Furthermore, we clarified that cotinine significantly facilitated tumor growth in the Lewis lung cancer model and accelerated development of lung adenomas induced by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone in A/J mice. We herein propose that cotinine induces tumor promotion by inhibiting apoptosis and enhancing cellular proliferation, thus underlining the importance of CYP2A6 in tobacco-related lung tumorigenesis.


Subject(s)
Apoptosis/drug effects , Cotinine/toxicity , Lung Neoplasms/pathology , Lung/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytochrome P-450 CYP2A6 , Female , Humans , Lung/cytology , Lung/pathology , Lung Neoplasms/genetics , Mice , Mice, Inbred A , Mice, Inbred C57BL , Nicotine/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
5.
Drug Metab Pharmacokinet ; 26(5): 451-7, 2011.
Article in English | MEDLINE | ID: mdl-21646728

ABSTRACT

Cytochrome P450 (CYP) 1A1 is involved in the metabolic activation of polycyclic aromatic hydrocarbons (PAHs) and is induced by several compounds, including PAHs. The induction of CYP1A1 mediated by the aryl hydrocarbon receptor (AhR) has been well investigated; however, little has been reported on the mechanisms of CYP1A1 induction mediated by factors other than AhR. In this study, we investigated the involvement of liver X receptor alpha (LXRα) in the induction of CYP1A1. TO-901317, an LXRα ligand, induced CYP1A1 mRNA in a dose-dependent fashion. Luciferase reporter assays using HepG2 cells showed that TO-901317 was capable of activating the promoter of the CYP1A1 gene and that a direct repeat 4 (DR4) motif located in a region from -452 to -467 was required for the induction of CYP1A1 through LXRα. Specific binding of LXRα to this DR4 motif was confirmed by gel shift and chromatin immunoprecipitation assays. Co-treatment of HepG2 cells with TO-901317 and 2,3,7,8-tetrachlorodibenzo-p-dioxin, a typical AhR ligand, caused the synergistic induction of CYP1A1 mRNA. Thus, we propose that the expression of CYP1A1 is regulated by LXRα as well as by AhR, suggesting that exposure to both LXRα and AhR ligands can result in the alteration of individual susceptibility to environmental carcinogens metabolically activated by CYP1A1.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Orphan Nuclear Receptors/physiology , Receptors, Aryl Hydrocarbon/physiology , Amino Acid Motifs/physiology , Hep G2 Cells , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver X Receptors , Polychlorinated Dibenzodioxins/pharmacology , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/drug effects , Sulfonamides/pharmacology
6.
Clin Chim Acta ; 412(13-14): 1249-51, 2011 Jun 11.
Article in English | MEDLINE | ID: mdl-21420395

ABSTRACT

BACKGROUND: Genetic polymorphisms of the human CYP2A6 gene are considered to be a determinant of smoking behavior and tobacco-related lung cancer risk in male Japanese smokers. We developed a SmartAmp-based genotyping method to detect whole deletion of the CYP2A6 gene directly from blood samples without DNA isolation. METHODS: We validated the new method using CYP2A plasmids, 48 genomic DNA samples and 25 blood samples by utilizing the SmartAmp method, a unique isothermal DNA amplification process. RESULTS: This method could discriminate the CYP2A6 gene from highly homologous CYP2A7 and CYP2A13 genes. CYP2A6*1 (wild-type) and CYP2A6*4 (whole gene deletion) were determined by the new method in perfect accordance with sequence analysis data. CONCLUSIONS: A SmartAmp assay for genotyping the CYP2A6 gene was developed, and the reliability of the method was validated using the conventional PCR method.


Subject(s)
Aryl Hydrocarbon Hydroxylases/blood , Aryl Hydrocarbon Hydroxylases/genetics , Gene Deletion , Nucleic Acid Amplification Techniques/methods , Aryl Hydrocarbon Hydroxylases/deficiency , Cytochrome P-450 CYP2A6 , DNA/blood , DNA/genetics , DNA Primers/genetics , Genotype , Hot Temperature , Humans , Time Factors
7.
Eur J Clin Pharmacol ; 66(10): 1065-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20700584

ABSTRACT

PURPOSE: although CYP2D6*36 was thought to be one of the alleles causing the poor metabolizer phenotype, several in vitro studies clarified that the enzyme produced by CYP2D6*36 showed enzymatic activities. However, the effects of CYP2D6*36 in tandem with CYP2D6*10 on the in vivo CYP2D6 activity have been unclear. In this study, we investigated in vivo metabolic capacities of CYP2D6 among the subjects carrying different numbers of CYP2D6*36 in tandem with CYP2D6*10. METHODS: we measured the metabolic ratio (MR) of dextromethorphan in 98 subjects. We determined the CYP2D6 genotype of these subjects, including allelic copy number of CYP2D6*10 and CYP2D6*36 by direct sequencing, TaqMan assay, and real-time Invader assay. RESULTS: single copies of CYP2D6*10 and tandem duplication of CYP2D6*36-*10 alleles were found at frequencies of 8.7 and 32.7%, respectively. Median dextromethorphan MRs of the subjects carrying CYP2D6*10 and CYP2D6*36-*10 were not significantly different (P = 0.24). CONCLUSIONS: CYP2D6*36 in tandem with CYP2D6*10 plays a minor role in interindividual variation of dextromethorphan metabolism in vivo.


Subject(s)
Asian People/genetics , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Dextromethorphan/metabolism , Gene Frequency , Adult , Excitatory Amino Acid Antagonists/metabolism , Genotype , Humans , Japan , Male , Phenotype , Reference Values
8.
Int J Environ Res Public Health ; 7(7): 2953-64, 2010 07.
Article in English | MEDLINE | ID: mdl-20717551

ABSTRACT

Human biomonitoring of plasma and urinary levels of nicotine, cotinine, and 3'-hydroxycotinine was conducted after daily cigarette smoking in a population of 92 male Japanese smokers with a mean age of 37 years who had smoked an average of 23 cigarettes per day for 16 years. Members of the population were genotyped for the nicotine-metabolizing enzyme cytochrome P450 2A6 (CYP2A6). The mean levels of nicotine, the levels of its metabolites cotinine and 3'-hydroxycotinine, and the sum of these three levels in subjects one hour after smoking the first cigarette on the sampling day were 20.1, 158, 27.7, and 198 ng/mL in plasma and 846, 1,020, 1,010, and 2,870 ng/mL in urine under daily smoking conditions. Plasma levels of 3'-hydroxycotinine and urinary levels of nicotine and 3'-hydroxycotinine were dependent on the CYP2A6 phenotype group, which was estimated from the CYP2A6 genotypes of the subjects, including those with whole gene deletion. Plasma cotinine levels were significantly correlated with the number of cigarettes smoked on the day before sampling (r = 0.71), the average number of cigarettes smoked daily (r = 0.58), and the Brinkman index (daily cigarettes x years, r = 0.48) under the present conditions. The sum of nicotine, cotinine, and 3'-hydroxycotinine concentrations in plasma showed a similar relationship to that of the plasma cotinine levels. Urinary concentrations of cotinine and the sum of nicotine metabolite concentrations also showed significant correlations with the plasma levels and the previous day's and average cigarette consumption. The numbers of cigarettes smoked per day by two subjects with self-reported light smoking habits were predicted by measuring the urinary cotinine concentrations and using linear regression equations derived from above-mentioned data. These results indicate that biomonitoring of the urinary cotinine concentration is a good, easy-to-use marker for plasma levels of cotinine and the sum of nicotine metabolites in smokers independent of genetic polymorphism of CYP2A6.


Subject(s)
Cotinine/urine , Nicotine/metabolism , Smoking/urine , Adult , Aryl Hydrocarbon Hydroxylases/genetics , Asian People , Biomarkers/urine , Cotinine/analogs & derivatives , Cytochrome P-450 CYP2A6 , Environmental Monitoring , Genotype , Humans , Linear Models , Male , Polymorphism, Genetic
9.
Chem Res Toxicol ; 22(2): 323-31, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19138062

ABSTRACT

5-n-Butyl-7-(3,4,5-trimethoxybenzoylamino)pyrazolo[1,5-a]pyrimidine) (OT-7100) is a pyrazolopyrimidine derivative with potential analgesic effects. Exclusively limited elevations in serum levels of aspirate- and alanine-aminotransferase were abnormally observed in a clinical study, in contrast to no toxicological potential to experimental animals. The aim of this study was to clarify the mechanism responsible for species-specific hepatotoxicity of this model compound. OT-7100 was primarily metabolized to a carboxylic acid derivative and an amino derivative (5-n-butyl-pyrazolo[1,5-a]pyrimidine, M-5) by hydrolysis in humans and rats. In human liver, pyrazolo[1,5-a]pyrimidine derivative M-5 was further metabolized to mainly M-23OH (a C-3-position hydroxyl derivative, 3-hydroxy-5-n-butyl-pyrazolo[1,5-a]pyrimidine). Studies with recombinant cytochrome P450s (P450s), correlation analysis using a panel of human liver microsomes as well as immunoinhibition with anti-P450 antibodies collectively suggested that human liver microsomal P450 1A2 preferentially metabolized M-5 to predominantly M-23OH. Human liver microsomes were capable of activating M-5 to a covalently bound metabolite faster than rat liver microsomes: reduced glutathione prevented the bindings. A cysteine adduct derivative of M-23OH at the C-6-position was structurally confirmed. On the contrary, rat liver microsomal P450 1A2 could metabolize M-5 to equally M-23OH, M-22OH (a C-6-position hydroxyl derivative, 6-hydroxy-5-n-butyl-pyrazolo[1,5-a]pyrimidine), or an unknown metabolite. These results suggest that differences in the regiospecific metabolic function of human and rat P450 1A2 would be responsible for the human-specific metabolic activation of the primary metabolite of OT-7100 to a proximate form. It is presumed that hepatotoxicity associated with OT-7100 could be likely related to the formation of a human-specific reactive metabolite from M-23OH. OT-7100 activation by inducible P450 1A2 may therefore exhibit marked individual differences.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Cytochrome P-450 CYP1A2/metabolism , Pyrazoles/metabolism , Pyrimidines/metabolism , Animals , Chemical and Drug Induced Liver Injury/etiology , Cysteine/chemistry , Cytochrome P-450 CYP1A2 Inhibitors , Humans , Hydrolysis , Magnetic Resonance Spectroscopy , Microsomes, Liver/enzymology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Species Specificity
10.
Drug Metab Dispos ; 37(4): 699-701, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19158312

ABSTRACT

During the course of sequencing for the CYP2D6 gene, we found a novel single nucleotide polymorphism of g.3318G>A (E383K) associated with CYP2D6*10, termed as CYP2D6*72. We also found a g.1611T>A (F120I) in the CYP2D6*49, which was previously identified as a CYP2D6*10-associated allele in an independent Japanese population. To clarify the effects of these novel CYP2D6*10 haplotypes on the functions of CYP2D6, kinetic analysis for dextromethorphan O-demethylation was performed using the Escherichia coli expression system and human liver microsomes. The V(max)/K(m) values for dextromethorphan O-demethylation catalyzed by recombinant CYP2D6 forms encoded by CYP2D6*10, CYP2D6*49, and CYP2D6*72 were 3.0, 0.5, and 1.3%, respectively, compared with that catalyzed by CYP2D6.1. Liver microsomes from a human subject genotyped as CYP2D6*10/*49 also showed a reduced dextromethorphan O-demethylase activity. CYP2D6.49 formed a 7-hydroxydextromethorphan, with a roughly similar V(max)/K(m) value to that of O-demethylation. These results suggest that these two CYP2D6*10 haplotypes are possible causes of interindividual variation in the activities and the substrate specificity of CYP2D6.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , Haplotypes , Dextromethorphan/pharmacokinetics , Humans , Japan , Microsomes, Liver/enzymology , Phenotype
11.
Drug Metab Dispos ; 37(1): 14-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18824522

ABSTRACT

The monkey CYP2C76 gene does not correspond to any of the human CYP2C genes, and its enzyme is at least partly responsible for the species difference occasionally seen in drug metabolism between monkeys and humans. To establish a line and/or lines of monkeys that are expected to show metabolic patterns highly similar to humans, we set out to find monkeys that lacked CYP2C76 activity. By genetic screening of 73 monkeys and a database search of expressed sequence tags, we found a total of 10 nonsynonymous genetic variants in the coding region of CYP2C76, including a null genotype (c.449TG>A). Some of the variants were differently distributed between two animal groups originating from different geographical regions (Indochina and Indonesia). After screening 170 additional genomic samples, we identified a total of eight animals (six males and two females) that were heterozygous for c.449TG>A, which could be used for establishing a homozygous line. If the homozygotes show drug-metabolizing properties more similar to humans than wild-type monkeys, the homozygotes may serve as a better animal model for drug metabolism. The data presented in this article provide the essential genetic information to perform a successful study by using cynomolgus monkeys and present a possible tool to generate a better animal model for drug metabolism.


Subject(s)
Alleles , Cytochrome P-450 Enzyme System/genetics , Models, Animal , Pharmacokinetics , Animals , Macaca fascicularis , Polymerase Chain Reaction
12.
Drug Metab Dispos ; 37(3): 453-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19088266

ABSTRACT

Glutathione S-transferase (GST) is one of the most important phase II drug-metabolizing enzymes, catalyzing the conjugation of electrophilic substrates to glutathione. Unlike in humans, a surprisingly limited number of GST genes have been identified in monkeys. The identification of additional GST genes in this model system would prove to be advantageous, because monkeys remain an important predictor of drug effects and toxicities in humans during preclinical studies. In this study, we report the identification and characterization of the following six cDNAs in cynomolgus monkeys: mfGSTA1, mfGSTA2, mfGSTM5, mfMGST1, mfGSTO1, and mfGSTZ1. These cDNAs encode GSTs highly homologous (approximately 95%) to human GST cDNAs. Among these, the mfGSTA1, mfGSTM5, mfMGST1, mfGSTO1, and mfGSTZ1 cDNAs correspond to a single human GST counterpart, whereas the mfGSTA2 cDNA is highly similar to human GSTA1 and GSTA2 cDNAs. An analysis of tissue samples indicates that these GST genes are predominantly expressed in the liver along with some extrahepatic expression as determined by real-time reverse transcriptase-polymerase chain reaction. It is interesting to note that mfGSTA2 is significantly differentially expressed between males and females in the jejunum, where a striking 8-fold higher expression level is observed in males. These results suggest that a potential sex difference in the metabolism of drugs may be mediated by mfGSTA2. This also provides a basis for the investigation of sex-dependent drug metabolism in monkeys.


Subject(s)
Gene Expression , Glutathione Transferase/genetics , Sex Factors , Amino Acid Sequence , Animals , Female , Glutathione Transferase/chemistry , Humans , Macaca fascicularis , Male , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
13.
Mol Med Rep ; 2(4): 585-8, 2009.
Article in English | MEDLINE | ID: mdl-21475870

ABSTRACT

Previously, we demonstrated that 8-methoxypsoralen (methoxsalen), a potent human cytochrome P450 2A6 (CYP2A6) inhibitor, strongly suppresses lung adenoma induction by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in female A/J mice. In the present study, we examined the inhibitory effects of methoxsalen on the development of lung adenocarcinomas, as well as on adenomas and alveolar hyperplasia. Female A/J mice were treated with methoxsalen at doses of 12.5 or 1.25 mg/kg body weight, administered by stomach tube once daily for 3 days. One hour after the final treatment, NNK was injected i.p. at a dose of 2 mg/mouse. The experiments were terminated 52 weeks after the first methoxsalen treatment, and lung adenomas and adenocarcinomas were analyzed histopathologically. Pretreatment with methoxsalen significantly reduced the incidence of adenocarcinomas from 94.7 to 46.7% (12.5 mg/kg) and 44.4% (1.25 mg/kg), and their tumor multiplicity from 4.68 to 0.87 (12.5 mg/kg) and 0.61 (1.25 mg/kg) tumors/mouse. The tumor multiplicity of adenomas and adenocarcinomas in the methoxsalen-treated groups was significantly reduced from 12.47 to 5.67 (12.5 mg/kg) and 4.28 (1.25 mg/kg) tumors/mouse. Approximately 60% of the adenocarcinomas arose within adenomas. In comparing the methoxsalen + NNK and NNK alone groups, there was no significant difference in the frequency of such compound lesions, indicating that pretreatment with methoxsalen did not suppress the eventual progression of adenomas to adenocarcinomas. These results clearly demonstrate that methoxsalen, a potent human CYP2A6 inhibitor, inhibits not only lung adenoma but also adenocarcinoma development.

14.
Cancer Sci ; 99(5): 1049-54, 2008 May.
Article in English | MEDLINE | ID: mdl-18380793

ABSTRACT

S-1 is an oral anticancer agent composed of tegafur (FT), 5-chloro-2,4-dihydroxypyridine (CDHP), and potassium oxonate. CDHP is added to prevent degradation of 5-fluorouracil (5-FU) by inhibiting dihydropyrimidine dehydrogenase. CYP2A6 is involved in the biotransformation of FT to 5-FU. Thus, we prospectively analyzed the effects of the CYP2A6 genotype, plasma level of CDHP, and patient characteristics on the pharmacokinetic (PK) variability of FT and 5-FU. Fifty-four Japanese patients with metastatic or recurrent cancers who received S-1 were enrolled. The CYP2A6 polymorphisms (*4A, *7, and *9) with deficient or reduced activity were analyzed. All subjects were classified into three groups according to their CYP2A6 genotype: wild type (*1/*1), one-variant allele (*1/any), or two-variant alleles (combination other than *1). The PK of FT, 5-FU, and CDHP were measured on day 1 of treatment. Multivariate regression analysis revealed that oral clearance of FT was associated with the CYP2A6 genotype (analysis of variance [ANOVA], P = 0.000838). The oral clearance of FT seen in patients with the two-variant alleles was significantly lower than those in wild type and the one-variant allele (95% confidence intervals 0.75-2.41 and 0.41-1.82, respectively; Tukey-Kramer test). The area under the time-concentration curve (AUC) of 5-FU was significantly correlated with the AUC of CDHP (ANOVA, P = 0.00126). The AUC of 5-FU and CDHP were inversely correlated with creatinine clearance (ANOVA, P = 0.0164 and P = 0.000762, respectively). Although the CYP2A6 variants are the cause of the PK variability of FT, the AUC of CDHP affected by renal function is the key determinant of the variability in the PK of 5-FU.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/genetics , Fluorouracil/pharmacokinetics , Mixed Function Oxygenases/genetics , Oxonic Acid/pharmacokinetics , Pyridines/blood , Tegafur/pharmacokinetics , Adult , Aged , Aged, 80 and over , Aryl Hydrocarbon Hydroxylases/metabolism , Asian People , Cytochrome P-450 CYP2A6 , Drug Combinations , Female , Genotype , Humans , Male , Middle Aged , Mixed Function Oxygenases/metabolism , Neoplasms/drug therapy , Polymorphism, Genetic
15.
Cancer Sci ; 98(10): 1582-5, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17683511

ABSTRACT

CYP2A6 metabolizes various nitrosamines, such as those in the diet and in tobacco smoke, which have been implicated as risk factors for colorectal tumors. To determine whether changes in expression levels could contribute to their progression, we carried out immunohistochemistry for CYP2A6 in human colon tumors. Colon specimens (n = 53) were diagnosed as adenoma (n = 16), adenocarcinoma (n = 30) or carcinoma in or with adenoma (n = 7). Colon tumor cells showed cytoplasmic granular immunoreactivity for CYP2A6. Adenocarcinomas and adjacent mucosa showed similar highly elevated degrees of CYP2A6 expression, whereas carcinomas in or with adenoma and adenomas showed lesser increases. To further determine whether CYP2A6 mRNA was expressed at the same level as the CYP2A6 protein, we carried out in situ hybridization of CYP2A6 in two cases of adenocarcinoma. In situ hybridization for CYP2A6 revealed mRNA expression in adenocarcinoma cells. The data indicate that CYP2A6 may have important roles in human colorectal tumorigenesis and progression, so that it could be a candidate therapeutic and chemopreventive target for colorectal cancers.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Colorectal Neoplasms/metabolism , Mixed Function Oxygenases/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Aged , Aryl Hydrocarbon Hydroxylases/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cytochrome P-450 CYP2A6 , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , In Situ Hybridization , Lymphatic Metastasis/pathology , Male , Mixed Function Oxygenases/genetics , Neoplasm Invasiveness/pathology , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Arch Biochem Biophys ; 466(1): 98-105, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17689485

ABSTRACT

Cynomolgus monkey CYP2C76 does not have a corresponding ortholog in humans, and it is at least partly responsible for differences in drug metabolism between monkeys and humans. To determine if CYP2C76 is the only monkey-specific CYP gene, we identified cynomolgus monkey cDNAs for CYP2A23, CYP2A24, CYP2E1, CYP2J2, CYP3A5, CYP3A8, CYP4A11, CYP4F3, CYP4F11, CYP4F12, and CYP4F45. These CYP cDNAs showed a high sequence identity (93-96%) to the homologous human CYP cDNAs. The monkey CYPs were preferentially expressed in liver among the analyzed tissues. Moreover, all five analyzed monkey CYPs (CYP2A23, CYP2A24, CYP2E1, CYP3A5, and CYP3A8) metabolized typical substrates for human CYPs in the corresponding subfamilies. These results suggest that these 11 monkey CYP cDNAs are closely related to the human CYP cDNAs and thus, unlike CYP2C76, are not apparent monkey-specific cDNAs.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/physiology , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/genetics , Humans , Macaca fascicularis , Molecular Sequence Data , Organ Specificity , Sequence Homology , Species Specificity , Tissue Distribution
17.
Oncol Rep ; 18(1): 53-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17549345

ABSTRACT

CYP2A6 is a major phase I enzyme metabolizing tobacco-specific nitrosamines, implicated as risk factors for lung cancer. In this study, immunohistochemistry and in situ hybridization (ISH) for CYP2A6 with human lung cancer tissues (n=31) obtained by surgical resection showed significantly higher immunoreactivity in the cases with lymph node metastasis. The adenocarcinoma cases (n=23) with lymph node metastasis or large tumor size showed a high immunoreactivity for CYP2A6. The squamous cell carcinoma cases (n=6) with large tumor size showed a tendency for low CYP2A6 immunoreactivity. ISH for CYP2A6 revealed mRNA expression in both adenocarcinoma and squamous cell carcinoma cells. The data suggest that CYP2A6 could have an important role in the development and proliferation of lung carcinomas. With adenocarcinomas, CYP2A6 could be a target candidate for therapeutic and chemopreventive intervention.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Lung Neoplasms/enzymology , Mixed Function Oxygenases/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/secondary , Aged , Carcinoma, Large Cell/enzymology , Carcinoma, Large Cell/secondary , Carcinoma, Small Cell/enzymology , Carcinoma, Small Cell/secondary , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/secondary , Cytochrome P-450 CYP2A6 , Disease Progression , Female , Humans , Immunoenzyme Techniques , Lung Neoplasms/pathology , Lymphatic Metastasis/pathology , Male , Neoplasm Invasiveness/pathology , Neoplasm Staging , Prognosis
18.
Nat Methods ; 4(3): 257-62, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17322893

ABSTRACT

We developed a rapid single nucleotide polymorphism (SNP) detection system named smart amplification process version 2 (SMAP 2). Because DNA amplification only occurred with a perfect primer match, amplification alone was sufficient to identify the target allele. To achieve the requisite fidelity to support this claim, we used two new and complementary approaches to suppress exponential background DNA amplification that resulted from mispriming events. SMAP 2 is isothermal and achieved SNP detection from whole human blood in 30 min when performed with a new DNA polymerase that was cloned and isolated from Alicyclobacillus acidocaldarius (Aac pol). Furthermore, to assist the scientific community in configuring SMAP 2 assays, we developed software specific for SMAP 2 primer design. With these new tools, a high-precision and rapid DNA amplification technology becomes available to aid in pharmacogenomic research and molecular-diagnostics applications.


Subject(s)
Base Pair Mismatch/genetics , DNA Mutational Analysis/methods , DNA Primers/genetics , Nucleic Acid Amplification Techniques/methods , Polymorphism, Single Nucleotide/genetics , Software , Suppression, Genetic/genetics , Algorithms , Base Sequence , Molecular Sequence Data , Sequence Analysis, DNA/methods , Temperature
19.
Mol Genet Metab ; 90(1): 58-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16996766

ABSTRACT

The reduced capacity of flavin-containing monooxygenase 3 (FMO3) to N-oxidize trimethylamine (TMA) is believed to cause a metabolic disorder. The aim of this study was to investigate the inter-individual variations of FMO3. Genomic DNA of case subjects that showed only 10-20% of FMO3 metabolic capacity among self-reported trimethylaminuria Japanese volunteers was sequenced. Functional analysis of recombinant FMO3 proteins was also performed. One homozygote for a novel single nucleotide substitution causing a stop codon at Arg500 was observed. The biological parents of this Proband A were heterozygous and showed >90% TMA N-oxygenation metabolic capacity. Another Proband B had the Arg500Stop and Cys197Stop codons. The TMA N-oxygenation metabolic capacities of the father and brother of this Proband B were apparently observed by possessing Arg205Cys mutant that coded for decreased TMA N-oxygenase. Recombinant Arg500Stop FMO3 cDNA expressed in Escherichia coli membranes and a series of highly purified truncation mutants at different positions of the C-terminus of FMO3 showed no detectable functional activity toward typical FMO3 substrates. The results suggest that individuals homozygous for either of the nonsense mutations, Arg500Stop and/or Cys197Stop alleles, in the FMO3 gene can possess abnormal TMA N-oxygenation.


Subject(s)
Codon, Nonsense/genetics , Methylamines/urine , Oxygenases/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Japan , Male , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/genetics , Methylamines/metabolism , Middle Aged , Oxidation-Reduction , Oxygenases/physiology , Pedigree
20.
Yakugaku Zasshi ; 126(12): 1309-18, 2006 Dec.
Article in Japanese | MEDLINE | ID: mdl-17139156

ABSTRACT

This paper introduces one of our projects performed at Hokkaido University. During the course of pharmacokinetic studies of SM-12502, which was under development as an anti-platelet-activating factor agent, we found three individuals who showed a slow metabolic phenotype in its pharmacokinetics. Analyzing the genes for CYP2A6 from the three, we discovered that they had the whole CYP2A6 gene deletion (CYP2A6*4C). Genetically engineered Salmonella YG7108 cells expressing human P450 were established to compare the mutagen-producing capacity of the P450 enzymes for various N-nitrosamines. We found that CYP2A6 was involved in the metabolic activation of N-nitrosamines with relatively bulky alkyl chains such as a tobacco-specific nitrosamine, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), which has been known to cause lung tumors in rodents. Thus, to examine the hypothesis that individuals possessing the CYP2A6*4C have a reduced risk of cancer due to the lack of the metabolic activation of certain carcinogens in tobacco smoke, a case-control study was performed. The results clearly indicated a significant association between the CYP2A6 genotype and lung cancer risk in smokers. In contrast, there was no significant relationship between them in nonsmokers. In addition, our results showed that the reduced risk of cancer was caused by the reduced activity of CYP2A6. Thus it was expected that the inhibition of the enzyme would result in a reduced cancer risk caused by smoking. The results of experiments using mice which were treated with NNK, a carcinogenic nitrosamine contained in tobacco smoke, together with 8-methoxypsolaren, a strong inhibitor of CYP2A6, indicated that the inhibition of CYP2A6 completely abolished the occurrence of adenoma.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Mixed Function Oxygenases , Molecular Biology , Toxicology , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/physiology , Cytochrome P-450 CYP2A6 , Gene Deletion , Humans , Lung Neoplasms/etiology , Lung Neoplasms/prevention & control , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/physiology , Nitrosamines/metabolism , Platelet Aggregation Inhibitors/pharmacokinetics , Polymorphism, Genetic , Risk , Smoking/adverse effects , Thiazolidines/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...