Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Mol Genet Metab ; 98(1-2): 198-202, 2009.
Article in English | MEDLINE | ID: mdl-19577495

ABSTRACT

Loss-of-function mutations in the flavin-containing monooxygenase 3 gene (FMO3) cause the inherited disorder trimethylaminuria (TMAuria), or fish-odour syndrome. Here we describe the identification in a family from northern Norway of a novel causative mutation of TMAuria. A female child within the family presented with a TMAuria-like phenotype. The child and her mother were found to be heterozygous for a novel mutation (R238Q) in exon 6 of FMO3. The child's father lacked this mutation, but was heterozygous for a double polymorphic variant, E158K/E308G, which was not present in the child. During a consultation with her doctor the mother mentioned an uncle whom she remembered as having a strong body odour. This discussion led to genetic counselling of the uncle and analysis of his DNA showed him to be homozygous for the R238Q mutation. Analysis of the mutant FMO3 expressed in bacteria revealed that the R238Q mutation abolished catalytic activity of the enzyme and is thus a causative mutation for TMAuria. The specificity constant (k(cat)/K(M)) of the K158/G308 variant was 43% of that of ancestral FMO3. Because the child is heterozygous for the R238Q mutation and no other mutation known to cause TMAuria was detected in her DNA she is predicted to suffer from transient childhood TMAuria, whereas her great-uncle has primary TMAuria.


Subject(s)
Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/genetics , Mutation/genetics , Oxygenases/genetics , White People/genetics , Alleles , Amino Acid Sequence , Base Sequence , Biocatalysis , DNA Mutational Analysis , Exons/genetics , Family , Female , Humans , Kinetics , Male , Molecular Sequence Data , Mutant Proteins/metabolism , Norway , Oxygenases/chemistry , Pedigree , Protein Structure, Secondary
2.
Biochem Pharmacol ; 62(6): 777-86, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11551524

ABSTRACT

The expression, in adult human skin, of genes encoding flavin-containing monooxygenases (FMOs) 1, 3, 4, and 5 and cytochromes P450 (CYPs) 2A6, 2B6, and 3A4 was determined by RNase protection. Each FMO and CYP exhibits inter-individual variation in expression in this organ. Of the individuals analysed, all contained CYP2B6 mRNA in their skin, 90% contained FMO5 mRNA and about half contained mRNAs encoding FMOs 1, 3, and 4, and CYPs 2A6 and 3A4. The amount of each of the FMO and CYP mRNAs in skin is much lower than in the organ in which it is most highly expressed, namely the kidney (for FMO1) and the liver (for the others). In contrast to the latter organs, in the skin FMO mRNAs are present in amounts similar to, or greater than, CYP mRNAs. Only the mRNA encoding CYP2B6 decreased in abundance in skin with increasing age of the individual. All of the mRNAs were substantially less abundant in cultures of keratinocytes than in samples of skin from which the cells were derived. In contrast, an immortalized human keratinocyte cell line, HaCaT, expressed FMO3, FMO5, and CYP2B6 mRNAs in amounts that fall within the range detected in the whole skin samples analysed. FMO1, CYP2A6, and CYP3A4 mRNAs were not detected in HaCaT cells, whereas FMO4 expression was markedly increased in this cell line compared to whole skin. In situ hybridization showed that the expression of each of the FMOs and CYPs analysed was localized to the epidermis, sebaceous glands and hair follicles.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Keratinocytes/enzymology , Oxygenases/metabolism , Skin/enzymology , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , Humans , Keratinocytes/metabolism , Oxygenases/genetics , RNA, Messenger/metabolism , Skin/cytology , Skin/metabolism , Subcellular Fractions
3.
Altern Lab Anim ; 29(3): 335-46, 2001.
Article in English | MEDLINE | ID: mdl-11387028

ABSTRACT

The addition of pyruvate to the culture medium has been reported to improve the maintenance of P450-dependent enzyme expression in primary rat hepatocyte cultures. In this study, the effects of 30mM pyruvate on cell morphology, albumin secretion and glutathione S-transferase (GST) expression were investigated as a function of the time in culture. The effect of triiodothyronine (T3) exposure on GST expression was also measured in pyruvate-treated cultures. Transmission electron microscopy showed that untreated hepatocytes deteriorated after culture for 7 days, whereas the morphology of the pyruvate-treated cells was similar to that observed in intact liver tissue. The albumin secretion rate was significantly higher in rat hepatocytes exposed to pyruvate than in control cells. In the presence of pyruvate, mu and alpha class GST activities were well maintained, whereas GST pi activity was increased over the entire culture period. HPLC analysis revealed that the complement of GST subunits present in hepatocytes is altered during culture with pyruvate: mu,class proteins remained relatively constant, whereas a decrease in the alpha class content was accompanied by a strong increase in GST subunit P1 (GSTP1). The induction of GSTP1 was confirmed at the mRNA level. In control cultures, pi class GST activity was increased, but total, mu, and alpha class GST activities continuously declined as a function of culture time and became undetectable beyond 7 days in culture. At the protein and mRNA levels, a much smaller increase in GSTP1 was observed than in the pyruvate cultures. When the pyruvate-treated cell cultures were exposed to T3, an inhibitory effect on GST activities and proteins was found. These results indicate that this simple culture model could be useful for studying the expression and regulation of GST.


Subject(s)
Glutathione Transferase/biosynthesis , Hepatocytes/drug effects , Hepatocytes/enzymology , Pyruvic Acid/pharmacology , Albumins/metabolism , Animals , Blotting, Northern , Cells, Cultured , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Hepatocytes/cytology , Isoenzymes/biosynthesis , Isoenzymes/genetics , Microscopy, Electron , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Triiodothyronine/pharmacology
4.
J Biol Chem ; 276(16): 12822-6, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11278292

ABSTRACT

The mechanisms by which different classes of chemicals induce the same cytochrome P450 (CYP) or the same chemical differentially induces more than one CYP are not well understood. We show that in primary hepatocytes and in vivo in liver (transfected by particle-mediated delivery) two orphan nuclear receptors, constitutive androstane receptor and pregnane X receptor (PXR1), transactivate a CYP gene via the same response element in a xenobiotic-specific manner. The constitutive androstane receptor mediates the barbiturate activation of expression of CYP2B1 and CYP3A1. PXR1 activates both genes in response to synthetic steroids. To exert their effect the receptors bind to the same direct repeat site (DR4) within the phenobarbital response element of the CYP2B1 promoter and to the same DR3 site in the pregnane X response element of CYP3A1. The receptors are therefore promiscuous with respect to DNA binding but not ligand binding. Differences in enhancer half-site spacing may influence the efficiency of interactions between the receptor and the transcription machinery and hence form the basis for the differential induction of CYP2B1 and CYP3A1 in response to barbiturates and synthetic steroids.


Subject(s)
Cytochrome P-450 CYP2B1/genetics , Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation, Enzymologic/physiology , Hepatocytes/enzymology , Mixed Function Oxygenases/genetics , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Transcription Factors/physiology , Xenobiotics/pharmacology , Animals , Biolistics , Cells, Cultured , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/genetics , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B1/biosynthesis , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/biosynthesis , Dimerization , Enzyme Induction/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/cytology , Mice , Mixed Function Oxygenases/biosynthesis , Pregnane X Receptor , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/physiology , Receptors, Steroid/genetics , Recombinant Fusion Proteins/biosynthesis , Retinoid X Receptors , Salamandridae , Transcription Factors/genetics , Transcriptional Activation , Transfection
5.
Biochem J ; 355(Pt 1): 71-8, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11256950

ABSTRACT

The constitutive androstane receptor (CAR) activates the expression of a reporter gene attached to the phenobarbital-response element (PBRE) of the cytochrome P450 2B1 (CYP2B1) gene in response to the barbiturate phenobarbital and the plant product picrotoxin. The xenobiotic-mediated increase in transactivation occurs in transfected primary hepatocytes and in liver transfected by biolistic-particle-mediated DNA transfer, but not in the transformed cell lines HepG2, CV-1 and HeLa, which support only constitutive activation of gene expression by CAR. Steroid co-activator 1 (SRC-1) enhances both constitutive and xenobiotic-induced CAR-mediated transactivation via the CYP2B1 PBRE in transfected primary hepatocytes. The nuclear receptor 1 (NR1) site of the PBRE is sufficient for CAR-mediated transactivation, but additional sequences within the PBRE, and hence the proteins that bind to them, are required for the interaction of CAR with SRC-1. The NR2 site of the PBRE binds proteins other than CAR, including an unidentified nuclear receptor heterodimerized with retinoid X receptor alpha. By binding to the proximal promoter of CYP2B1, the transcription factor Sp1 increases both basal transcription and xenobiotic-induced expression via the PBRE. Thus induction of CYP2B1 expression by xenobiotics is mediated by the nuclear receptor CAR and, for optimal expression, requires SRC-1 and Sp1.


Subject(s)
Cytochrome P-450 CYP2B1/biosynthesis , Receptors, Cytoplasmic and Nuclear/physiology , Sp1 Transcription Factor/physiology , Transcription Factors/physiology , Xenobiotics/pharmacology , Animals , Base Sequence , Cell Line, Transformed , Cells, Cultured , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B1/genetics , DNA Primers , Enzyme Induction/drug effects , Enzyme Induction/physiology , Hepatocytes/drug effects , Hepatocytes/enzymology , Histone Acetyltransferases , Nuclear Receptor Coactivator 1 , Rats , Transcriptional Activation/drug effects
6.
Biochem Pharmacol ; 60(10): 1491-6, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11020451

ABSTRACT

Ethanol, a human toxicant and a solvent in pharmacological research, is known to interfere with biotransformation of xenobiotics. We compared the in vivo and in vitro long-term effects of ethanol exposure on the expression of glutathione S-transferases (GST, EC 2. 5.1.18) in rat liver. Long-term in vivo ethanol treatment to achieve blood ethanol levels ranging between 10-50 mM was by liquid diet feeding. For in vitro experiments, rat hepatocytes co-cultured with rat liver epithelial cells were exposed to 17 and 68 mM ethanol for up to 10 days. Two weeks of liquid diet ethanol treatment increased total GST activity. Both Mu and Alpha classes and in particular the A1 and A2 subunits and the amount of their corresponding mRNAs were increased. Total GST activity was also increased in co-cultures after exposure to 68 mM ethanol for 10 days. However, the Mu class subunits M1 and M2 and the corresponding mRNAs were increased, rather than the Alpha class subunits. Thus, long-term exposure to ethanol induces hepatic GST both in vivo and in vitro, but different isoenzymes are affected. Consequently, extrapolation of in vitro data on GST expression and regulation to the in vivo situation must be judicious. During xenobiotic metabolism in cell culture, a shift in relative expression and induction of different GST forms may occur, resulting in either an under- or overestimation of effects.


Subject(s)
Ethanol/pharmacology , Glutathione Transferase/biosynthesis , Liver/drug effects , Animals , Biotransformation/drug effects , Enzyme Induction/drug effects , Glutathione Transferase/drug effects , Glutathione Transferase/genetics , In Vitro Techniques , Liver/enzymology , Liver/metabolism , Male , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley
7.
Toxicol Appl Pharmacol ; 168(3): 216-24, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11042094

ABSTRACT

The flavin-containing monooxygenases (FMOs) are a family of xenobiotic-metabolizing enzymes that are expressed in a species- and tissue-specific manner. FMO2 expression has been observed in pulmonary tissue from several species, but not human. Two human FMO2 point mutations have been reported: a cytosine to thymidine transition at position 1414 resulting in a premature stop codon and a thymidine insertion at position 1589 resulting in a frameshift. To define the frequency of these sequence variations and explore their significance, unrelated African-American, Caucasian, and Korean individuals were genotyped. In the African-American population tested (n = 180), the 1414C allele occurred at a 13% frequency; however, all of the tested Caucasians (n = 52) and Koreans (n = 100) were homozygous for the 1414T allele. The T1589 allele occurred at frequencies of 6.9 and 13.0% in African-Americans (n = 175) and Caucasians (n = 23), respectively, and appears to segregate with the 1414T allele. Thus, it would have no further impact on FMO2 activity. Western blot analysis of pulmonary microsomes failed to detect immunoreactive protein in 1414T homozygotes. A heterozygotic individual did exhibit a single band of the expected size, but no detectable FMO activity in the corresponding lung microsomes. Sequence analysis, however, was consistent with the 1414C allele encoding an active FMO2 enzyme. FMO2 mRNA expression was observed in most individuals, but failed to correlate with genotype or protein expression. In summary, functional FMO2 is expressed in only a small percentage of the overall population. However, in certain ethnic groups, active pulmonary FMO2 enzyme will be present in a significant number of individuals.


Subject(s)
Black People/genetics , Oxygenases/genetics , Polymorphism, Genetic/genetics , Alleles , Blotting, Western , Genotype , Humans , Oligonucleotides/analysis , Oligonucleotides/genetics , Oxygenases/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , United States
8.
J Endocrinol ; 166(2): 363-71, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10927625

ABSTRACT

Glutathione S-transferases (GSTs) are subject to regulation by thyroid and sex hormones and by GH. We have used an in vitro experimental system comprising adult rat hepatocytes co-cultured with rat liver epithelial cells of primitive biliary origin, to distinguish between direct and indirect effects of various hormones on GSTs; to identify the GST subunits affected by individual hormones; and to investigate the level at which the hormones act. Tri-iodothyronine (T3), thyroxine (T4) and 17beta-oestradiol (OE2) reduced GST activities, whereas testosterone, dihydrotestosterone, and human growth hormone (hGH) had little effect on total GST activity. HPLC separation of the various GST subunits revealed that T3 and T4 reduced total GST content, in particular the abundance of subunits M1 and M2. The amount of the Pi-class subunit P1 was reduced by OE2. Treatment of the co-cultured cells with this hormone altered the GST subunit profile to one that is more similar to that observed in freshly isolated hepatocytes. Analysis of mRNAs demonstrated that some of the hormones act at a pre-translational level, whereas others act at a translational or post-translational level to regulate the expression of various GST subunits.


Subject(s)
Glutathione Transferase/metabolism , Gonadal Steroid Hormones/pharmacology , Hepatocytes/enzymology , Human Growth Hormone/pharmacology , Isoenzymes/metabolism , Thyroid Hormones/pharmacology , Animals , Blotting, Northern , Cells, Cultured , Coculture Techniques , Dihydrotestosterone/pharmacology , Epithelial Cells/enzymology , Estradiol/pharmacology , Gene Expression Regulation , Glutathione Transferase/genetics , Hepatocytes/drug effects , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Testosterone/pharmacology , Thyroxine/pharmacology , Triiodothyronine/pharmacology
9.
Pharmacogenetics ; 10(5): 439-51, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10898113

ABSTRACT

We have previously shown that primary trimethylaminuria, or fish-odour syndrome, is caused by an inherited defect in the flavin-containing monooxygenase 3 (FMO3) catalysed N-oxidation of the dietary-derived malodorous amine, trimethylamine (TMA). We now report a novel causative mutation for the disorder identified in a young girl diagnosed by proton nuclear magnetic resonance (NMR) spectroscopy of her urine. Sequence analysis of genomic DNA amplified from the patient revealed that she was homozygous for a T to C missense mutation in exon 3 of the FMO3 gene. The mutation changes an ATG triplet, encoding methionine, at codon 82 to an ACG triplet, encoding threonine. A polymerase chain reaction/restriction enzyme-based assay was devised to genotype individuals for the FMO3Thr82 allele. Wild-type and mutant FMO3, heterologously expressed in a baculovirus-insect cell system, were assayed by ultraviolet spectrophotometry and NMR spectroscopy for their ability to catalyse the N-oxidation of TMA. The latter technique has the advantage of enabling the simultaneous, direct and semi-continuous measurement of both of the products, TMA N-oxide and NADP, and of one of the reactants, NADPH. Results obtained from both techniques demonstrate that the Met82Thr mutation abolishes the catalytic activity of the enzyme and thus represents the genetic basis of the disorder in this individual. The combination of NMR spectroscopy with gene sequence and expression technology provides a powerful means of determining genotype-phenotype relationships in trimethylaminuria.


Subject(s)
Genetic Diseases, Inborn/enzymology , Genetic Diseases, Inborn/genetics , Mutation/genetics , Odorants , Oxygenases/genetics , Adult , Alleles , Amino Acid Sequence , Amino Acid Substitution/genetics , Base Sequence , Child, Preschool , Female , Genetic Diseases, Inborn/urine , Genotype , Humans , Infant , Methylamines/blood , Methylamines/urine , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Oxygenases/analysis , Oxygenases/biosynthesis , Sequence Analysis, DNA , Syndrome , Threonine/genetics
10.
Pharmacogenetics ; 10(9): 799-807, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11191884

ABSTRACT

Fish-odour syndrome is a highly unpleasant disorder of hepatic trimethylamine (TMA) metabolism characterized by a body odour reminiscent of rotting fish, due to excessive excretion of the malodorous free amine. Although fish-odour syndrome may exhibit as sequelae with other conditions (e.g. liver dysfunction), many patients exhibit an inherited, more persistent form of the disease. Ordinarily, dietary-derived TMA is oxidized to the nonodorous N-oxide by hepatic flavin-containing monooxygenase 3 (FMO3). Our previous demonstration that a mutation, P153L (C to T), in the FMO3 gene segregated with the disorder and inactivated the enzyme confirmed that defects in FMO3 underlie the inherited form of fish-odour syndrome. We have investigated the genetic basis of the disorder in two further affected pedigrees and report that the three propositi are all compound heterozygotes for causative mutations of FMO3. Two of these individuals possess the P153L (C to T) mutation and a novel mutation, N61S (A to G). The third is heterozygous for novel, M4341 (G to A), and previously reported, R492W (C to T), mutations. Functional characterization of the S61, 1434 and W492 variants, via baculovirus-mediated expression in insect cells, confirmed that all three mutations either abolished, or severely attenuated, the capacity of the enzyme to catalyse TMA N-oxidation. Although 1434 and W492 were also incapable of catalysing the S-oxidation of methimazole, S61 was fully active with this sulphur-containing substrate. Since an asparagine is conserved at the equivalent position to N61 of FMO3 in mammalian, yeast and Caenorhabditis elegans FMOs, the characterization of the naturally occurring N61S (A to G) mutation may have identified this asparagine as playing a critical role specifically in FMO-catalysed N-oxidation.


Subject(s)
Flavoproteins/genetics , Metabolism, Inborn Errors/genetics , Methylamines/urine , Mutation, Missense/genetics , Oxygenases/genetics , Base Sequence , Female , Gene Frequency , Genotype , Heterozygote , Humans , Male , Molecular Sequence Data , Odorants , Pedigree , Recombinant Proteins , Syndrome
11.
J Biol Chem ; 273(46): 30599-607, 1998 Nov 13.
Article in English | MEDLINE | ID: mdl-9804831

ABSTRACT

Flavin-containing monooxygenases (FMOs) are NADPH-dependent flavoenzymes that catalyze the oxidation of heteroatom centers in numerous drugs and xenobiotics. FMO2, or "pulmonary" FMO, one of five forms of the enzyme identified in mammals, is expressed predominantly in lung and differs from other FMOs in that it can catalyze the N-oxidation of certain primary alkylamines. We describe here the isolation and characterization of cDNAs for human FMO2. Analysis of the sequence of the cDNAs and of a section of the corresponding gene revealed that the major FMO2 allele of humans encodes a polypeptide that, compared with the orthologous protein of other mammals, lacks 64 amino acid residues from its C terminus. Heterologous expression of the cDNA revealed that the truncated polypeptide was catalytically inactive. The nonsense mutation that gave rise to the truncated polypeptide, a C --> T transition in codon 472, is not present in the FMO2 gene of closely related primates, including gorilla and chimpanzee, and must therefore have arisen in the human lineage after the divergence of the Homo and Pan clades. Possible mechanisms for the fixation of the mutation in the human population and the potential significance of the loss of functional FMO2 in humans are discussed.


Subject(s)
Oxygenases/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Catalysis , Codon, Nonsense , Codon, Terminator , Humans , Macaca fascicularis , Molecular Sequence Data , Pan troglodytes , RNA, Messenger/metabolism , Ribonucleases/metabolism
12.
Biochem Pharmacol ; 56(8): 1047-51, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9776317

ABSTRACT

To investigate the hormonal control of the expression of flavin-containing monooxygenase (FMO; EC 1.14.13.8) under defined in vitro conditions, adult male rat hepatocytes were isolated by collagenase perfusion and co-cultured with rat liver epithelial cells of primitive biliary origin. The direct effect of 17beta-estradiol, testosterone, 5alpha-dihydrotestosterone (5alpha-DHT) and human growth hormone (hGH) on FMO activity was studied using this in vitro model. Optimal, non-cytotoxic hormonal concentrations were determined by measuring the lactate dehydrogenase (LDH) index. In addition, the microsomal protein content of the cultured hepatocytes was determined as a function of culture time. The female sex hormone 17beta-estradiol caused a significant decrease in FMO as a function of culture time. After 14 days of exposure, FMO activity decreased by 56%. Neither of the male sex hormones or human growth hormone had an effect on FMO activity. These results in co-cultured male rat hepatocytes support in vivo observation that 17beta-estradiol is a potent hormone involved in the negative regulation of the expression of FMO in male rat liver.


Subject(s)
Gonadal Steroid Hormones/pharmacology , Human Growth Hormone/pharmacology , Liver/drug effects , Microsomes, Liver/drug effects , Oxygenases/metabolism , Steroids/pharmacology , Animals , Coculture Techniques , Dihydrotestosterone/pharmacology , Epithelial Cells/drug effects , Estradiol/pharmacology , Liver/cytology , Liver/enzymology , Male , Microsomes, Liver/enzymology , Rats , Rats, Sprague-Dawley , Sexual Maturation/physiology , Testosterone/pharmacology
13.
Toxicol In Vitro ; 12(4): 335-41, 1998 Aug.
Article in English | MEDLINE | ID: mdl-20654415

ABSTRACT

The regulation of flavin-containing monooxygenase (FMO) by thyroid hormones was examined under well defined in vitro conditions using adult male rat hepatocytes co-cultured with rat liver epithelial cells of primitive biliary origin. Serum free medium was used to avoid interferences from foetal bovine serum. The effect of thyroxine (T4) and its major metabolite l-triiodothyronine (T3) on FMO activity was estimated spectrophotometrically by measuring the rate of methimazole oxygenation. The highest non-cytotoxic doses of T3 and T4 that could be used in co-cultures were determined by measuring both lactate dehydrogenase leakage into the medium and microsomal protein content of the hepatocytes as a function of culture time. In addition, hormonal responsiveness of the in vitro system used was confirmed by malic enzyme activity measurements. Administration of 10 mum T3 or T4 was found to cause a significant decrease in FMO activity and content, suggesting a suppressive role of both hormones on the regulation of FMO activity in male rats.

15.
Nat Genet ; 17(4): 491-4, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9398858

ABSTRACT

Individuals with primary trimethylaminuria exhibit a body odour reminiscent of rotting fish, due to excessive excretion of trimethylamine (TMA; refs 1-3). The disorder, colloquially known as fish-odour syndrome, is inherited recessively as a defect in hepatic N-oxidation of dietary-derived TMA and cannot be considered benign, as sufferers may display a variety of psychosocial reactions, ranging from social isolation of clinical depression and attempted suicide. TMA oxidation is catalyzed by flavin-containing mono-oxygenase (FMO; refs 7,8), and tissue localization and functional studies have established FMO3 as the form most likely to be defective in fish-odour syndrome. Direct sequencing of the coding exons of FMO3 amplified from a patient with fish-odour syndrome identified two missense mutations. Although one of these represented a common polymorphism, the other, a C-->T transition in exon 4, was found only in an affected pedigree, in which it segregated with the disorder. The latter mutation predicts a proline-->leucine substitution at residue 153 and abolishes FMO3 catalytic activity. Our results indicate that defects in FMO3 underlie fish-odour syndrome and that the Pro 153-->Leu 153 mutation described here is a cause of this distressing condition.


Subject(s)
Metabolism, Inborn Errors/enzymology , Methylamines/urine , Mutation , Odorants , Oxygenases/genetics , Amino Acid Sequence , Animals , Base Sequence , Humans , Metabolism, Inborn Errors/genetics , Molecular Sequence Data , Oxidation-Reduction , Pedigree , Syndrome
17.
Genomics ; 46(2): 260-7, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9417913

ABSTRACT

The inherited metabolic disorder trimethylaminuria (fish-odor syndrome) is associated with defective hepatic N-oxidation of dietary-derived trimethylamine catalyzed by flavin-containing monooxygenase (FMO). As FMO3 encodes the major form of FMO expressed in adult human liver, it represents the best candidate gene for the disorder. The structural organization of FMO3 was determined by sequencing the products of exon-to-exon and vectorette PCR, the latter through the use of vectorette libraries constructed directly from genomic DNA. The gene contains one noncoding and eight coding exons. Knowledge of the exon/intron organization of the human FMO3 gene enabled each of the coding exons of the gene, together with their associated flanking intron sequences, to be amplified from genomic DNA and will thus facilitate the identification of mutations in FMO3 in families affected with fish-odor syndrome.


Subject(s)
Metabolism, Inborn Errors/genetics , Methylamines/metabolism , Oxygenases/genetics , Exons , Flavin-Adenine Dinucleotide/metabolism , Humans , Introns , Molecular Sequence Data , NADP/metabolism , Odorants , Oxygenases/metabolism , Polymerase Chain Reaction/methods , Sequence Analysis, DNA
18.
Genomics ; 34(3): 426-9, 1996 Jun 15.
Article in English | MEDLINE | ID: mdl-8786146

ABSTRACT

The human flavin-containing monooxygenase (FMO) gene family comprises at least five distinct members (FMO1 to FMO5) that code for enzymes responsible for the oxidation of a wide variety of soft nucleophilic substrates, including drugs and environmental pollutants. Three of these genes (FMO1, FMO3, and FMO4) have previously been localized to human chromosome 1q, raising the possibility that the entire gene family is clustered in this chromosomal region. Analysis by polymerase chain reaction of DNA isolated from a panel of human-rodent somatic cell hybrids demonstrates that the two remaining identified members of the FMO gene family, FMO2 and FMO5, also are located on chromosome 1q.


Subject(s)
Chromosomes, Human, Pair 1 , Multigene Family , Oxygenases/genetics , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , Codon , Cricetinae , DNA Primers , DNA, Complementary , Humans , Hybrid Cells , Liver/enzymology , Molecular Sequence Data , Oxygenases/metabolism , Polymerase Chain Reaction , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Substrate Specificity
19.
Eur J Biochem ; 235(3): 683-9, 1996 Feb 01.
Article in English | MEDLINE | ID: mdl-8654418

ABSTRACT

We have previously described the isolation and sequencing of cDNA clones encoding flavin-containing monooxygenases (FMOs) 1 and 4 of man [Dolphin, C., Shephard, E. A., Povey, S., Palmer, C. N. A., Ziegler, D. M., Ayesh, R., Smith, R. L. & Phillips, I. R. (1991) J. Biol. Chem. 266, 12379-12385; Dolphin, C., Shephard E. A., Povey, S., Smith, R. L. & Phillips, I. R. (1992) Biochem. J. 287, 261-267]. We present here the isolation of a cDNA for FM03 of man. The sequence of this CDNA and the amino acid sequence deduced from it differ substantially from those previously reported for this member of the FMO family of man. In addition, we have investigated, by quantitative RNase protection assays, the expression in several foetal and adult human tissues of genes encoding FMO1, FMO3 and FMO4, Our results demonstrate that, in the adult, FMO1 is expressed in kidney but not in liver, whereas in the foetus it is expressed in both organs. The lack of expression of FMO1 in adult human liver is in marked contrast to the situation in other mammals, such as pig and rabbit, in which FMO1 constitutes a major form of the enzyme in the liver of the adult animal. The mRNA encoding FMO3 is abundant in adult liver and is also present, in low abundance, in some foetal tissues. Thus, FMO1 and FMO3 are both subject to developmental and tissue-specific regulation, with a developmental switch in the expression of the genes taking place in the liver. FMO4 mRNA is present in low abundance in several foetal and adult tissues and thus the corresponding gene appears to be expressed constitutively.


Subject(s)
Gene Expression Regulation, Developmental , Oxygenases/genetics , Adult , Amino Acid Sequence , Base Sequence , DNA, Complementary , Humans , Liver/embryology , Liver/enzymology , Molecular Sequence Data , RNA, Messenger/genetics
20.
Eur J Biochem ; 231(2): 337-43, 1995 Jul 15.
Article in English | MEDLINE | ID: mdl-7635145

ABSTRACT

It has been shown previously that the anticonvulsant agent, sodium valproate, induces certain cytochrome P-450 monooxygenase activities and decreases glutathione S-transferase activity. We have used Western blotting, RNase protection assays and Northern blot hybridization to determine the effects of valproate on the abundance of individual components of the cytochrome P-450 monooxygenase and of glutathione S-transferase subunits. Due to the short half-life of the drug in rats we have used an in vitro experimental system comprised of rat hepatocytes co-cultured with rat primitive biliary epithelial cells. Valproate was shown to be a potent inducer of two members of the cytochrome P-450 (CYP)2B subfamily, CYP2B1 and 2B2. The induction of the proteins was mediated at the level of the mRNAs, with the mRNA for CYP2B1 being more highly induced than that for CYP2B2. The drug also induced, but to a much lesser extent, two important components of the cytochrome-P-450-mediated monooxygenase system, NADPH-dependent cytochrome P-450 reductase and cytochrome b5, and their corresponding mRNAs. Thus, the effects of valproate on cytochromes P-450 and other components of the cytochrome-P450-mediated monooxygenase system mimic those of another, structurally diverse, antiepileptic drug, phenobarbital. However, in contrast to phenobarbital, which induces glutathione S-transferase subunits 1, 2, 3, 4 and 7, valproate selectively decreases the abundance of subunits 3 and/or 4. It has been shown previously that CYP2B1 is involved in the production of metabolites of valproate implicated in hepatotoxicity. The induction of this protein by valproate would thus contribute substantially to the hepatotoxic effects associated with the drug.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Glutathione Transferase/biosynthesis , Microsomes, Liver/drug effects , Steroid Hydroxylases/biosynthesis , Valproic Acid/pharmacology , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , Cytochromes b5/biosynthesis , Cytochromes b5/genetics , Enzyme Induction , Glutathione Transferase/genetics , Male , Microsomes, Liver/enzymology , NADPH-Ferrihemoprotein Reductase , Oxidoreductases/biosynthesis , Oxidoreductases/genetics , Phenobarbital/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Ribonucleases/metabolism , Steroid Hydroxylases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...