Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Anim Sci ; 89(12): 4032-42, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21803974

RESUMEN

The cervix and its secretions undergo biochemical and physical changes under the differential influences of estrogen and progesterone. These include changes in the glycoprotein profile of the endocervix and its secretions. A comprehensive survey of such changes in cervical epithelium and cervical secretions was performed on bovine samples throughout the periestrous period. Cervical tissue samples and swabs were collected from synchronized beef heifers that were slaughtered 1) 12 h after controlled intravaginal progesterone-releasing device (CIDR) removal, 2) 24 h after CIDR removal, 3) at the onset of estrus, 4) 12 h after the onset of estrus, 5) 48 h after the onset of estrus, and 6) 7 d after the onset of estrus. Histological staining with hematoxylin and eosin, periodic acid Schiff, Alcian blue, and high-iron diamine was carried out to map overall patterns of stored glycoproteins and tissue structure. Biotinylated lectins were also used to detect the presence and distribution of a range of saccharide structures. The activities of ß-galactosidase, α-L-fucosidase, ß-N-acetyl-hexosaminidase, and sialidase were measured in cervical swabs using specific substrates. The epithelial layer of the cervix exhibited dynamic changes in cellular hypertrophy and amounts of stored glycoprotein. The greatest content of neutral and acidic mucins was observed 48 h after onset of estrus (P < 0.05). Sialylated mucins predominated at the bases of cervical folds, whereas sulfated mucins were more abundant (P < 0.05) at their apices. The stained area of core mucin glycans changed (P < 0.05) in association with follicular versus luteal phases, whereas terminal glycans changed (P < 0.05) mainly at the time of estrus and shortly thereafter. The greatest activity of ß-galactosidase and sialidase was observed 12 h after onset of estrus, whereas ß-hexosaminidase and α-fucosidase peaked at the luteal time point (P < 0.05). Taken together, we suggest that the well-known changes in the endocervix and its secretions that are associated with the physiological modulation of sperm transport and function of the cervical barrier are, in part, driven by glycosylation changes.


Asunto(s)
Bovinos/fisiología , Cuello del Útero/metabolismo , Estro/fisiología , Glicoproteínas/metabolismo , Glicósido Hidrolasas/metabolismo , Animales , Epitelio/metabolismo , Femenino , Glicoproteínas/genética , Glicósido Hidrolasas/genética , Lectinas/metabolismo , Mucinas/fisiología , Unión Proteica
3.
Biochem Pharmacol ; 62(6): 777-86, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11551524

RESUMEN

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.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Queratinocitos/enzimología , Oxigenasas/metabolismo , Piel/enzimología , Células Cultivadas , Sistema Enzimático del Citocromo P-450/genética , Humanos , Queratinocitos/metabolismo , Oxigenasas/genética , ARN Mensajero/metabolismo , Piel/citología , Piel/metabolismo , Fracciones Subcelulares
4.
Toxicol Appl Pharmacol ; 168(3): 216-24, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11042094

RESUMEN

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.


Asunto(s)
Población Negra/genética , Oxigenasas/genética , Polimorfismo Genético/genética , Alelos , Western Blotting , Genotipo , Humanos , Oligonucleótidos/análisis , Oligonucleótidos/genética , Oxigenasas/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estados Unidos
5.
Pharmacogenetics ; 10(5): 439-51, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10898113

RESUMEN

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.


Asunto(s)
Enfermedades Genéticas Congénitas/enzimología , Enfermedades Genéticas Congénitas/genética , Mutación/genética , Odorantes , Oxigenasas/genética , Adulto , Alelos , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Secuencia de Bases , Preescolar , Femenino , Enfermedades Genéticas Congénitas/orina , Genotipo , Humanos , Lactante , Metilaminas/sangre , Metilaminas/orina , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Oxigenasas/análisis , Oxigenasas/biosíntesis , Análisis de Secuencia de ADN , Síndrome , Treonina/genética
6.
Pharmacogenetics ; 10(9): 799-807, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11191884

RESUMEN

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.


Asunto(s)
Flavoproteínas/genética , Errores Innatos del Metabolismo/genética , Metilaminas/orina , Mutación Missense/genética , Oxigenasas/genética , Secuencia de Bases , Femenino , Frecuencia de los Genes , Genotipo , Heterocigoto , Humanos , Masculino , Datos de Secuencia Molecular , Odorantes , Linaje , Proteínas Recombinantes , Síndrome
8.
J Biol Chem ; 273(46): 30599-607, 1998 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-9804831

RESUMEN

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.


Asunto(s)
Oxigenasas/genética , Alelos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Catálisis , Codón sin Sentido , Codón de Terminación , Humanos , Macaca fascicularis , Datos de Secuencia Molecular , Pan troglodytes , ARN Mensajero/metabolismo , Ribonucleasas/metabolismo
9.
J Neurochem ; 70(4): 1366-75, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9523552

RESUMEN

We report the isolation, by RT-PCR, of partial cDNAs encoding the rat peroxisome proliferator-activated receptor (PPAR) isoforms PPAR alpha, PPAR beta, and PPAR gamma and the rat retinoid X receptor (RXR) isoforms RXR alpha, RXR beta, and RXR gamma. These cDNAs were used to generate antisense RNA probes to permit analysis, by the highly sensitive and discriminatory RNase protection assay, of the corresponding mRNAs in rat brain regions during development. PPAR alpha, PPAR beta, RXR alpha, and RXR beta mRNAs are ubiquitously present in different brain regions during development, PPAR gamma mRNA is essentially undetectable, and RXR gamma mRNA is principally localised to cortex. We demonstrate, for the first time, the presence of PPAR and RXR mRNAs in primary cultures of neonatal meningeal fibroblasts, cerebellar granule neurons (CGNs), and cortical and cerebellar astrocytes and in primary cultures of adult cortical astrocytes. PPAR alpha, PPAR beta, RXR alpha, and RXR beta mRNAs are present in all cell types, albeit that PPAR alpha and RXR alpha mRNAs are at levels near the limit of detection in CGNs. PPAR gamma mRNA is expressed at low levels in most cell types but is present at levels similar to those of PPAR alpha mRNA in adult astrocytes. RXR gamma mRNA is present either at low levels, or below the level of detection of the assay, for all cell types studied.


Asunto(s)
Encéfalo/metabolismo , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Ácido Retinoico/genética , Factores de Transcripción/genética , Animales , Células Cultivadas , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Femenino , Isomerismo , Meninges/citología , Meninges/metabolismo , Plásmidos/genética , Ratas , Ratas Wistar , Receptores X Retinoide , Distribución Tisular
10.
Biochem J ; 329 ( Pt 2): 373-81, 1998 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9425122

RESUMEN

We have investigated, by RNase protection assays in rat brain regions and primary cortical astrocyte cultures, the presence of the mRNA species encoding the three mitochondrially located enzymes acetoacetyl-CoA thiolase, mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase (mt. HMG-CoA synthase) and HMG-CoA lyase (HMG-CoA lyase) that together constitute the ketogenic HMG-CoA cycle. As a prerequisite we obtained a full-length cDNA encoding rat HMG-CoA lyase by degenerate oligonucleotide-primed PCR coupled to a modification of PCR-rapid amplification of cDNA ends (PCR-RACE). We report here: (1) the nucleotide sequence of rat mt. HMG-CoA lyase, (2) detection of the mRNA species encoding all three HMG-CoA cycle enzymes in all regions of rat brain during suckling, (3) approximately twice the abundance of mt. HMG-CoA synthase mRNA in cerebellum than in cortex in 11-day-old suckling rat pups, (4) significantly lower abundances of mt. HMG-CoA synthase mRNA in brain regions derived from rats weaned to a high-carbohydrate/low-fat diet compared with the corresponding regions derived from the suckling rat, and (5) the presence of mt. HMG-CoA synthase mRNA in primary cultures of neonatal cortical astrocytes at an abundance similar to that found in liver of weaned animals. These results provide preliminary evidence that certain neural cell types possess ketogenic potential and might thus have a direct role in the provision of fatty acid-derived ketone bodies during the suckling period.


Asunto(s)
Acilcoenzima A/metabolismo , Sistema Nervioso Central/enzimología , Oxo-Ácido-Liasas/genética , ARN Mensajero/genética , Secuencia de Aminoácidos , Animales , Animales Lactantes , Astrocitos/enzimología , Astrocitos/metabolismo , Secuencia de Bases , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Clonación Molecular , ADN Complementario , Femenino , Datos de Secuencia Molecular , Plásmidos , Ratas , Ratas Wistar , Homología de Secuencia de Aminoácido , Destete
11.
Nat Genet ; 17(4): 491-4, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9398858

RESUMEN

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.


Asunto(s)
Errores Innatos del Metabolismo/enzimología , Metilaminas/orina , Mutación , Odorantes , Oxigenasas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Humanos , Errores Innatos del Metabolismo/genética , Datos de Secuencia Molecular , Oxidación-Reducción , Linaje , Síndrome
12.
Genomics ; 46(2): 260-7, 1997 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9417913

RESUMEN

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.


Asunto(s)
Errores Innatos del Metabolismo/genética , Metilaminas/metabolismo , Oxigenasas/genética , Exones , Flavina-Adenina Dinucleótido/metabolismo , Humanos , Intrones , Datos de Secuencia Molecular , NADP/metabolismo , Odorantes , Oxigenasas/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Análisis de Secuencia de ADN
13.
Genomics ; 34(3): 426-9, 1996 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8786146

RESUMEN

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.


Asunto(s)
Cromosomas Humanos Par 1 , Familia de Multigenes , Oxigenasas/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Clonación Molecular , Codón , Cricetinae , Cartilla de ADN , ADN Complementario , Humanos , Células Híbridas , Hígado/enzimología , Datos de Secuencia Molecular , Oxigenasas/metabolismo , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
14.
Eur J Biochem ; 235(3): 683-9, 1996 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-8654418

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Oxigenasas/genética , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , ADN Complementario , Humanos , Hígado/embriología , Hígado/enzimología , Datos de Secuencia Molecular , ARN Mensajero/genética
15.
Chem Biol Interact ; 96(1): 17-32, 1995 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-7720101

RESUMEN

cDNA clones encoding five distinct members of the FMO family of man (FMOs 1, 2, 3, 4 and 5) were isolated by a combination of library screening and reverse transcription-polymerase chain reaction techniques. The deduced amino acid sequences of the human FMOs have 82-87% identity with their known orthologues in other mammal but only 51-57% similarity to each other. The hydropathy profiles of the proteins are very similar. From the calculated rate of evolution of FMOs (a 1% change in sequence per 6 million years) it would appear that individual members of the FMO gene family arose by duplication of a common ancestral gene some 250-300 million years ago. Each of the FMO genes was mapped by the polymerase chain reaction to the long arm of human chromosome 1. The localization of the FMO1 gene was further refined to 1q23-q25 by in situ hybridization of human metaphase chromosomes. RNase protection assays demonstrated that in man each FMO gene displays a distinct developmental and tissue-specific pattern of expression. In the adult, FMO1 is expressed in kidney but not in liver, whereas in the foetus its mRNA is abundant in both organs. FMO3 expression is essentially restricted to the liver in the adult and the mRNA is either absent, or present in low amounts, in foetal tissues. FMO4 is expressed more constitutively. Human FMO1 and FMO3 cDNAs were functionally expressed in prokaryotic and eukaryotic cells. FMO1 and FMO3, expressed in either system, displayed product stereoselectivity in their catalysis of the N-oxidation of the pro-chiral tertiary amines, N-ethyl-N-methylaniline (EMA) and pargyline. Both enzymes were stereoselective with respect to the production of the (-)-S-enantiomer of EMA N-oxide. But in the case of pargyline, the enzymes displayed opposite stereoselectivity, FMO1 producing solely the (+)-enantiomer and FMO3 predominantly the (-)-enantiomer of the N-oxide.


Asunto(s)
ADN Complementario/aislamiento & purificación , Regulación Enzimológica de la Expresión Génica/genética , Oxigenasas/genética , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Cromosomas Humanos Par 1/genética , Clonación Molecular , ADN Complementario/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Biblioteca de Genes , Humanos , Hibridación in Situ , Datos de Secuencia Molecular , Peso Molecular , Oxigenasas/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Estándares de Referencia , Homología de Secuencia de Aminoácido , Translocación Genética
16.
Arch Biochem Biophys ; 308(1): 254-7, 1994 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8311461

RESUMEN

A nomenclature based on comparisons of amino acid sequences is proposed for the members of the mammalian flavin-containing monooxygenase (FMO) gene family. This nomenclature is based on evidence of a single gene family composed of five genes. The percentage identities of the amino acid sequences of the five known forms of mammalian FMO are between 52 and 57% in rabbit and between 50 and 58% across species lines. The identities of all orthologs are greater than 82%. There is no evidence for multiple, highly related forms of the enzyme or for more than one mammalian FMO gene family. In the proposed system, the mammalian flavin-containing monooxygenase gene family is designated as "FMO" and the individual genes are distinguished by an Arabic numeral. The FMOs known as the "liver" and "lung" enzymes become FMO1 and FMO2, and the more recently described forms of the enzymes become FMO3, FMO4, and FMO5. Human FMO gene designations, FMO1 and FMO3, remain unchanged, but the gene designated FMO2 becomes FMO4. Following convention, the genes and cDNA designations will be italicized and the mRNA and protein designations will be nonitalicized. The purpose of the proposed nomenclature is to provide for the unambiguous identification of orthologous forms of mammalian FMOs, regardless of the species or tissue in question. The proposed classification considers only members of the mammalian flavin-containing monooxygenase gene family and has no bearing on the generally accepted definition of a multisubstrate flavin-containing monooxygenase.


Asunto(s)
Secuencia de Aminoácidos , Isoenzimas/genética , Mamíferos/genética , Familia de Multigenes , Oxigenasas/genética , Animales , Cobayas , Humanos , Isoenzimas/química , Hígado/enzimología , Pulmón/enzimología , Oxigenasas/química , Conejos , Ratas , Homología de Secuencia de Aminoácido , Porcinos , Terminología como Asunto
17.
Genomics ; 16(1): 85-9, 1993 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8486388

RESUMEN

We have used the polymerase chain reaction to map the gene encoding human flavin-containing monooxygenase (FMO) form II (N. Lomri, Q. Gu, and J. R. Cashman, 1992, Proc. Natl. Acad. Sci. USA 89: 1685-1689) to chromosome 1. We propose the designation FMO3 for this gene as it is the third FMO gene to be mapped. The two other human FMO genes identified to date, FMO1 and FMO2, are also located on chromosome 1 (C. Dolphin, E. A. Shephard, S. Povey, C. N. A. Palmer, D. M. Ziegler, R. Ayesh, R. L. Smith, and I. R. Phillips, 1991, J. Biol. Chem. 266: 12379-12385; C. Dolphin, E. A. Shephard, S. F. Povey, R. L. Smith, and I. R. Phillips, 1992, Biochem. J. 286: 261-267). The localization of FMO1, FMO2, and FMO3 has been refined to the long arm of chromosome 1. Analysis of human metaphase chromosomes by in situ hybridization confirmed the mapping of FMO1 and localized this gene more precisely to 1q23-q25.


Asunto(s)
Cromosomas Humanos Par 1 , Oxigenasas/genética , Secuencia de Bases , Mapeo Cromosómico , ADN/genética , Humanos , Hibridación in Situ , Datos de Secuencia Molecular , Familia de Multigenes , Reacción en Cadena de la Polimerasa
18.
Biochem J ; 287 ( Pt 1): 261-7, 1992 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-1417778

RESUMEN

We have previously reported the cloning of cDNAs for a flavin-containing mono-oxygenase (FMO) of man, designated FMO1 [Dolphin, Shephard, Povey, Palmer, Ziegler, Ayesh, Smith & Phillips (1991) J. Biol. Chem. 266, 12379-12385], that is the orthologue of pig and rabbit hepatic FMOs. We now describe the isolation and characterization of cDNA clones for a second human FMO, which we have designated FMO2. The polypeptide encoded by the cDNAs is 558 amino acid residues long, has a calculated M(r) of 63337, and contains putative FAD- and NADP-binding sites that align exactly with those described in other mammalian FMOs. Human FMO2 has 51-53% primary sequence identity with human FMO1, rabbit pulmonary FMO and rabbit liver FMO form 2, and thus represents a fourth, distinct, member of the mammalian FMO family. The corresponding mRNA is present in low abundance in adult human liver. Southern blot hybridization with single-exon probes demonstrated that human FMO2 and FMO1 are the products of single genes. The gene encoding FMO2 (designated FMO2) was mapped, by the polymerase chain reaction, to human chromosome 1, the same chromosome on which FMO1 is located.


Asunto(s)
Oxigenasas/genética , Secuencia de Aminoácidos , Secuencia de Bases , Cromosomas Humanos Par 1 , Clonación Molecular , ADN/genética , Genes , Humanos , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Mapeo Restrictivo , Alineación de Secuencia , Solubilidad
19.
J Biol Chem ; 266(19): 12379-85, 1991 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-1712018

RESUMEN

cDNA clones that code for a pig and human flavin-containing monooxygenase (FMO) have been isolated. The full-length sequence of the human cDNAs revealed that they encode a polypeptide of 532 amino acid residues containing putative FAD- and NADP-binding sites. The deduced amino acid sequence has 88 and 86% identity, respectively, with the pig and rabbit "hepatic" forms of FMO, but is only 58% similar to the rabbit "pulmonary" FMO, and thus represents the human ortholog of the "hepatic" form of FMO. However, as this FMO is present in low abundance in human adult liver, the general term "hepatic" for this form of the enzyme is misleading, and thus we propose the name FMO1 to describe this human FMO and its mammalian orthologs. Northern blot analysis demonstrated that human FMO1 mRNA is more abundant in fetal than in adult liver, indicating that in man the enzyme is subject to developmental regulation. Southern blot hybridization of human genomic DNA suggests that the protein is encoded by a single gene, which has been designated FMO1 and mapped to chromosome 1.


Asunto(s)
Mapeo Cromosómico , Oxigenasas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Southern Blotting , Cromosomas Humanos Par 1 , Clonación Molecular , ADN/genética , Humanos , Células Híbridas , Hígado/enzimología , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN/genética , Conejos , Homología de Secuencia de Ácido Nucleico , Porcinos
20.
Am J Hum Genet ; 43(3): 280-4, 1988 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-2901225

RESUMEN

CYP3, the gene which encodes the hepatic cytochrome P450pcn1, the isozyme responsible for the metabolic oxidation of the calcium channel-blocking drug nifedipine, has recently been mapped to human chromosome 7 using somatic cell hybrids. Using multilocus linkage analysis in CEPH families, we examined the linkage of a cDNA probe (hPCN1) for CYP3 to the oncogene MET, the pro-alpha 2(1) collagen gene COL1A2, and the T-cell receptor beta-chain gene TCRB, together with three arbitrary loci D7S8, D7S13, and D7S16, defined by the anonymous DNA probes pJ3.11, pB79a, and p7C22, respectively. From 70 CEPH parents screened with a StyI RFLP for hPCN1, four informative families were found each with both parental and maternal grandparents and 6-11 children per family. Tight linkage emerged between CYP3 and COL1A2, with a maximum combined lod score of 5.72 at theta = 0, suggesting the most likely subchromosomal localization of CYP3 is 7q21.3-q22.1.


Asunto(s)
Cromosomas Humanos Par 7 , Colágeno/genética , Sistema Enzimático del Citocromo P-450/genética , Oxigenasas de Función Mixta/genética , Clonación Molecular , Citocromo P-450 CYP3A , Femenino , Ligamiento Genético , Humanos , Masculino , Nifedipino , Linaje , Polimorfismo de Longitud del Fragmento de Restricción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...