Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diabetologia ; 56(8): 1813-25, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23680914

RESUMO

AIMS/HYPOTHESIS: Mutations in BSCL2/seipin cause Berardinelli-Seip congenital lipodystrophy (BSCL), a rare recessive disorder characterised by near absence of adipose tissue and severe insulin resistance. We aimed to determine how seipin deficiency alters glucose and lipid homeostasis and whether thiazolidinediones can rescue the phenotype. METHODS: Bscl2 (-/-) mice were generated and phenotyped. Mouse embryonic fibroblasts (MEFs) were used as a model of adipocyte differentiation. RESULTS: As observed in humans, Bscl2 (-/-) mice displayed an early depletion of adipose tissue, with insulin resistance and severe hepatic steatosis. However, Bscl2 (-/-) mice exhibited an unexpected hypotriglyceridaemia due to increased clearance of triacylglycerol-rich lipoproteins (TRL) and uptake of fatty acids by the liver, with reduced basal energy expenditure. In vitro experiments with MEFs demonstrated that seipin deficiency led to impaired late adipocyte differentiation and increased basal lipolysis. Thiazolidinediones were able to rescue the adipogenesis impairment but not the alteration in lipolysis in Bscl2 (-/-) MEFs. In vivo treatment of Bscl2 (-/-) mice with pioglitazone for 9 weeks increased residual inguinal and mesenteric fat pads as well as plasma leptin and adiponectin concentrations. Pioglitazone treatment increased energy expenditure and improved insulin resistance, hypotriglyceridaemia and liver steatosis in these mice. CONCLUSIONS/INTERPRETATION: Seipin plays a key role in the differentiation and storage capacity of adipocytes, and affects glucose and lipid homeostasis. The hypotriglyceridaemia observed in Bscl2 (-/-) mice is linked to increased uptake of TRL by the liver, offering a new model of liver steatosis. The demonstration that the metabolic complications associated with BSCL can be partially rescued with pioglitazone treatment opens an interesting therapeutic perspective for BSCL patients.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/deficiência , Tiazolidinedionas/uso terapêutico , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Células Cultivadas , Metabolismo Energético/fisiologia , Feminino , Subunidades gama da Proteína de Ligação ao GTP , Proteínas Heterotriméricas de Ligação ao GTP/genética , Lipodistrofia Generalizada Congênita/tratamento farmacológico , Lipodistrofia Generalizada Congênita/metabolismo , Camundongos , Camundongos Mutantes , Pioglitazona , Gravidez
2.
Atherosclerosis ; 212(1): 246-51, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20619837

RESUMO

OBJECTIVE: Proprotein convertase subtilisin kexin/type 9 (PCSK9) is an inhibitor of the low density (LDL) lipoprotein receptor. Plasma PCSK9 is increased by fenofibrate and statins. Here, we determined how standard dose of statin and combined therapy with fenofibrate modulate PCSK9. METHODS: Randomized, open-label cross-over study investigating the effect of fenofibrate (160 mg), atorvastatin (10 mg), and combination of both in patients with type 2 diabetes mellitus and atherogenic dyslipidemia. After the single administration of atorvastatin and fenofibrate for 6 weeks, patients received both for another 6 weeks. PCSK9, lipids and lipoproteins levels were determined at day 1, weeks 6, 9 and 12. RESULTS: Upon 6 weeks of treatment, atorvastatin decreased LDL-cholesterol by 30% (p<0.001) and fenofibrate decreased triglyceride level by 31% (p<0.01) and increased HDL-cholesterol by 13% (p<0.05). Combination did not show further benefit. Atorvastatin increased PCSK9 by 24% at day 1 and by 14% at week 6 (p < or = 0.01). Fenofibrate increased PCSK9 by 26% at week 6 (p < or = 0.01), but had no effect at day 1. Three weeks of combination therapy increased PCSK9 by 42%, 6 weeks by 19% (p < or = 0.01). PCSK9 changes were not different between treatments over 6-week periods. CONCLUSION: Fenofibrate and atorvastatin increased circulating PCSK9 in diabetic patients, with no additive effect after 6 weeks of combined therapy.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Dislipidemias/tratamento farmacológico , Fenofibrato/uso terapêutico , Ácidos Heptanoicos/uso terapêutico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hipolipemiantes/uso terapêutico , Pirróis/uso terapêutico , Serina Endopeptidases/sangue , Adulto , Idoso , Atorvastatina , Biomarcadores/sangue , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Estudos Cross-Over , Diabetes Mellitus Tipo 2/sangue , Quimioterapia Combinada , Dislipidemias/sangue , Feminino , França , Alemanha , Humanos , Masculino , Pessoa de Meia-Idade , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Fatores de Tempo , Resultado do Tratamento , Triglicerídeos/sangue , Regulação para Cima
3.
Genomics ; 87(1): 84-92, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16314073

RESUMO

Congenital erythropoietic porphyria (CEP) is a recessive autosomal disorder characterized by a deficiency in uroporphyrinogen III synthase (UROS), the fourth enzyme of the heme biosynthetic pathway. The severity of the disease, the lack of specific treatment except for allogeneic bone marrow transplantation, and the knowledge of the molecular lesions are strong arguments for gene therapy. An animal model of CEP has been designed to evaluate the feasibility of retroviral gene transfer in hematopoietic stem cells. We have previously demonstrated that the knockout of the Uros gene is lethal in mice (Uros(del) model). This work describes the achievement of a knock-in model, which reproduces a mutation of the UROS gene responsible for a severe UROS deficiency in humans (P248Q missense mutant). Homozygous mice display erythrodontia, moderate photosensitivity, hepatosplenomegaly, and hemolytic anemia. Uroporphyrin (99% type I isomer) accumulates in urine. Total porphyrins are increased in erythrocytes and feces, while Uros enzymatic activity is below 1% of the normal level in the different tissues analyzed. These pathological findings closely mimic the CEP disease in humans and demonstrate that the Uros(mut248) mouse represents a suitable model of the human disease for pathophysiological, pharmaceutical, and therapeutic purposes.


Assuntos
Substituição de Aminoácidos , Mutação de Sentido Incorreto , Porfiria Eritropoética/enzimologia , Uroporfirinogênio III Sintetase/genética , Animais , Transplante de Medula Óssea , Modelos Animais de Doenças , Terapia Genética , Camundongos , Camundongos Transgênicos , Porfiria Eritropoética/patologia , Porfiria Eritropoética/terapia , Uroporfirinogênio III Sintetase/metabolismo , Uroporfirinas/metabolismo
4.
Mol Ther ; 4(4): 331-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11592836

RESUMO

Successful treatment of blood disorders by gene therapy has several complications, one of which is the frequent lack of selective advantage of genetically corrected cells. Erythropoietic protoporphyria (EPP), caused by a ferrochelatase deficiency, is a good model of hematological genetic disorders with a lack of spontaneous in vivo selection. This disease is characterized by accumulation of protoporphyrin in red blood cells, bone marrow, and other organs, resulting in severe skin photosensitivity. Here we develop a self-inactivating lentiviral vector containing human ferrochelatase cDNA driven by the human ankyrin-1/beta-globin HS-40 chimeric erythroid promoter/enhancer. We collected bone marrow cells from EPP male donor mice for lentiviral transduction and injected them into lethally irradiated female EPP recipient mice. We observed a high transduction efficiency of hematopoietic stem cells resulting in effective gene therapy of primary and secondary recipient EPP mice without any selectable system. Skin photosensitivity was corrected for all secondary engrafted mice and was associated with specific ferrochelatase expression in the erythroid lineage. An erythroid-specific expression was sufficient to reverse most of the clinical and biological manifestations of the disease. This improvement in the efficiency of gene transfer with lentiviruses may contribute to the development of successful clinical protocols for erythropoietic diseases.


Assuntos
Células da Medula Óssea/metabolismo , Modelos Animais de Doenças , Terapia Genética/métodos , Lentivirus/genética , Porfiria Hepatoeritropoética/genética , Porfiria Hepatoeritropoética/terapia , Animais , Southern Blotting , Transplante de Medula Óssea , Linhagem Celular , Elementos Facilitadores Genéticos/genética , Feminino , Ferroquelatase/genética , Ferroquelatase/metabolismo , Ferroquelatase/uso terapêutico , Expressão Gênica/genética , Vetores Genéticos/genética , Humanos , Lentivirus/fisiologia , Masculino , Camundongos , Especificidade de Órgãos , Porfiria Hepatoeritropoética/enzimologia , Porfiria Hepatoeritropoética/patologia , Porfirinas/metabolismo , Regiões Promotoras Genéticas/genética , Protoporfiria Eritropoética , Pele/patologia , Transdução Genética
5.
Biochem J ; 358(Pt 1): 257-62, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11485575

RESUMO

A cytosolic glutathione S-transferase (GST, EC 2.5.1.18) from the recently characterized Omega class [GSTO; Board et al. 2000, J. Biol. Chem. 275, 24798-24806] has been identified in pig organs. It was found widely distributed in the different tissues investigated and especially abundant in liver and muscle. The hepatic enzyme has been purified to homogeneity by using its selective affinity for S-hexylglutathione over GSH, thus providing a simple method to isolate mammalian GSTO. The dimeric protein has a subunit molecular mass of 27328 Da as measured by electrospray ionization MS. Internal peptide sequencing and complete cDNA sequencing revealed strong similarities with its human recombinant orthologue and two rodent GST-like proteins with the ability to catalyse the GSH-dependent reduction of dehydroascorbate. Additional similarities, including the presence of a specific N-terminal extension and of immunological cross-reactivity, support the results. Moreover, this gene encoding GSTO generates two organ-specific transcripts, suggesting transcriptional mechanisms with a significance that is as yet uncharacterized.


Assuntos
Glutationa Transferase/química , Fígado/enzimologia , Músculos/enzimologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Western Blotting , Cromatografia Líquida de Alta Pressão , DNA Complementar/metabolismo , Glutationa/análogos & derivados , Glutationa/química , Humanos , Immunoblotting , Camundongos , Dados de Sequência Molecular , Peptídeos/química , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Espectrometria de Massas por Ionização por Electrospray , Frações Subcelulares , Suínos , Fatores de Tempo , Distribuição Tecidual
6.
Biochem J ; 355(Pt 2): 481-8, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11284737

RESUMO

Liver fatty-acid-binding protein (L-FABP) is a cytoplasmic polypeptide that binds with strong affinity especially to long-chain fatty acids (LCFAs). It is highly expressed in both the liver and small intestine, where it is thought to have an essential role in the control of the cellular fatty acid (FA) flux. Because expression of the gene encoding L-FABP is increased by both fibrate hypolipidaemic drugs and LCFAs, it seems to be under the control of transcription factors, termed peroxisome-proliferator-activated receptors (PPARs), activated by fibrate or FAs. However, the precise molecular mechanism by which these regulations take place remain to be fully substantiated. Using transfection assays, we found that the different PPAR subtypes (alpha, gamma and delta) are able to mediate the up-regulation by FAs of the gene encoding L-FABP in vitro. Through analysis of LCFA- and fibrate-mediated effects on L-FABP mRNA levels in wild-type and PPARalpha-null mice, we have found that PPARalpha in the intestine does not constitute a dominant regulator of L-FABP gene expression, in contrast with what is known in the liver. Only the PPARdelta/alpha agonist GW2433 is able to up-regulate the gene encoding L-FABP in the intestine of PPARalpha-null mice. These findings demonstrate that PPARdelta can act as a fibrate/FA-activated receptor in tissues in which it is highly expressed and that L-FABP is a PPARdelta target gene in the small intestine. We propose that PPARdelta contributes to metabolic adaptation of the small intestine to changes in the lipid content of the diet.


Assuntos
Proteínas de Transporte/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Ácido Linoleico/farmacologia , Fígado/efeitos dos fármacos , Proteínas de Neoplasias , Proteínas do Tecido Nervoso , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Bezafibrato/farmacologia , Butiratos/farmacologia , Primers do DNA , Proteína 7 de Ligação a Ácidos Graxos , Proteínas de Ligação a Ácido Graxo , Genes Reporter , Hipolipemiantes , Intestino Delgado/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Compostos de Fenilureia/farmacologia , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Receptores Citoplasmáticos e Nucleares/agonistas , Fatores de Transcrição/agonistas , Transcrição Gênica/fisiologia
8.
Exp Eye Res ; 71(4): 395-404, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10995560

RESUMO

Fibroblast growth factors (FGF) are multipotent cytokines with demonstrated mitogenic, neurotrophic and angiogenic properties. There is evidence that they have multiple functions during and after development of the vertebrate eye. Amongst these, the role of FGF receptor mediated signaling in the retinal pigmented epithelium (RPE) is not yet well understood. FGF-2 is produced in RPE cells and may play a role in photoreceptor development and/or survival in vivo. It may also stimulate growth of melanocytes and angiogenesis in the choroid. To address these questions, we have specifically disrupted FGF signaling by generating lines of transgenic mice expressing dominant-negative FGF receptor 1 (FGFR-1) in the pigmented cells. Histological analysis of the eyes were conducted on hemizygous and homozygous mice at different ages. In homozygotes, eye growth is strongly impaired during embryogenesis leading to massive eye degeneration seen in the early post-natal stages. In hemizygotes, the choroid is thinned and the finger-like junctions between RPE cells and photoreceptors are disrupted. Scanning electron microscopy of the choroid vasculature showed that choriocapillary density, diameter and branching are strongly affected. As mice age, they develop progressive retinal degeneration as evidenced by photoreceptor cell loss. Our results are in agreement with the hypothesis that FGF signaling in the RPE participates in photoreceptor survival in vivo. Our model provides evidence that FGF signaling is also involved in choroidal angiogenesis by a process that could relate to induction of terminal branching.


Assuntos
Anormalidades do Olho/genética , Epitélio Pigmentado Ocular/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Animais , Comunicação Celular/fisiologia , Sobrevivência Celular , Olho/irrigação sanguínea , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Neovascularização Fisiológica , Células Fotorreceptoras de Vertebrados/fisiologia , Epitélio Pigmentado Ocular/citologia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia
9.
J Biol Chem ; 275(42): 33053-8, 2000 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-10918065

RESUMO

Mutations of the ABC1 transporter have been identified as the defect in Tangier disease, characterized by low HDL and cholesterol ester accumulation in macrophages. A full-length mouse ABC1 cDNA was used to investigate the mechanisms of lipid efflux to apoA-I or HDL in transfected 293 cells. ABC1 expression markedly increased cellular cholesterol and phospholipid efflux to apoA-I but had only minor effects on lipid efflux to HDL. The increased lipid efflux appears to involve a direct interaction between apoA-I and ABC1, because ABC1 expression substantially increased apoA-I binding at the cell surface, and chemical cross-linking and immunoprecipitation analysis showed that apoA-I binds directly to ABC1. In contrast to scavenger receptor BI (SR-BI), another cell surface molecule capable of facilitating cholesterol efflux, ABC1 preferentially bound lipid-free apoA-I but not HDL. Immunofluorescence confocal microscopy showed that ABC1 is primarily localized on the cell surface. In the absence of apoA-I, cells overexpressing ABC1 displayed a distinctive morphology, characterized by plasma membrane protrusions and resembling echinocytes that form when there are excess lipids in the outer membrane hemileaflet. The studies provide evidence for a direct interaction between ABC1 and apoA-I, but not HDL, indicating that free apoA-I is the metabolic substrate for ABC1. Plasma membrane ABC1 may act as a phospholipid/cholesterol flippase, providing lipid to bound apoA-I, or to the outer membrane hemileaflet.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Apolipoproteína A-I/metabolismo , Membrana Celular/fisiologia , Membrana Celular/ultraestrutura , Colesterol/metabolismo , Glicoproteínas/metabolismo , Proteínas de Membrana , Receptores de Lipoproteínas , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Sítios de Ligação , Antígenos CD36 , Linhagem Celular , Reagentes de Ligações Cruzadas , Glicoproteínas/genética , Humanos , Cinética , Lipoproteínas HDL/metabolismo , Camundongos , Microscopia Confocal , Fosfolipídeos/metabolismo , Receptores Imunológicos/fisiologia , Receptores Depuradores , Proteínas Recombinantes/metabolismo , Receptores Depuradores Classe B , Especificidade por Substrato , Transfecção
10.
J Biol Chem ; 275(36): 28240-5, 2000 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-10858438

RESUMO

Tangier disease, a condition characterized by low levels of high density lipoprotein and cholesterol accumulation in macrophages, is caused by mutations in the ATP-binding cassette transporter ABC1. In cultured macrophages, ABC1 mRNA was induced in an additive fashion by 22(R)-hydroxycholesterol and 9-cis-retinoic acid (9CRA), suggesting induction by nuclear hormone receptors of the liver X receptor (LXR) and retinoid X receptor (RXR) family. We cloned the 5'-end of the human ABC1 transcript from cholesterol-loaded THP1 macrophages. When transfected into RAW macrophages, the upstream promoter was induced 7-fold by 22(R)-hydroxycholesterol, 8-fold by 9CRA, and 37-fold by 9CRA and 22(R)-hydroxycholesterol. Furthermore, promoter activity was increased in a sterol-responsive fashion when cotransfected with LXRalpha/RXR or LXRbeta/RXR. Further experiments identified a direct repeat spaced by four nucleotides (from -70 to -55 base pairs) as a binding site for LXRalpha/RXR or LXRbeta/RXR. Mutations in this element abolished the sterol-mediated activation of the promoter. The results show sterol-dependent transactivation of the ABC1 promoter by LXR/RXR and suggest that small molecule agonists of LXR could be useful drugs to reverse foam cell formation and atherogenesis.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Glicoproteínas/genética , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores do Ácido Retinoico/metabolismo , Esteróis/farmacologia , Fatores de Transcrição/metabolismo , Ativação Transcricional , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/química , Alitretinoína , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Proteínas de Ligação a DNA , Biblioteca Gênica , Glicoproteínas/química , Humanos , Hidroxicolesteróis/farmacologia , Fígado/metabolismo , Receptores X do Fígado , Macrófagos , Dados de Sequência Molecular , Receptores Nucleares Órfãos , Receptores Citoplasmáticos e Nucleares/genética , Receptores do Ácido Retinoico/genética , Proteínas Recombinantes/metabolismo , Receptores X de Retinoides , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Doença de Tangier/genética , Fatores de Transcrição/genética , Transcrição Gênica , Ativação Transcricional/efeitos dos fármacos , Transfecção , Tretinoína/farmacologia , Células Tumorais Cultivadas
11.
Toxicol Lett ; 111(3): 243-51, 2000 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-10643869

RESUMO

Although numerous studies report strong hepatic cytochrome P450 decrease during aflatoxicosis, the mechanisms involved in this decrease remain to be established. The purpose of this work is to investigate whether decreased CYP mRNA expression could explain decreased P450 expression and activity. Studies were conducted in primary cultures of rabbit hepatocytes exposed to 0.1 and 1 microM aflatoxin B1 (AFB1) incubated in the culture medium for 72 h. In order to confirm the effects of the mycotoxin, 30 microM beta-naphthoflavone or rifampicin were used as respective inductors of P450 1A1 and 1A2 or 3A6. Dose-dependent decreases of CYP mRNA expression were observed in all AFB1-treated cells; however, these decreases were not specific. Moreover, P450 expression and activity are far less decreased by the AFB1 treatment than their corresponding mRNA. Taken together, these results suggest that the specific P450 decrease observed during aflatoxicosis was not the consequence of a specific decrease of their mRNA expression.


Assuntos
Aflatoxina B1/farmacologia , Hidrocarboneto de Aril Hidroxilases , Citocromo P-450 CYP1A1/biossíntese , Citocromo P-450 CYP1A2/biossíntese , Sistema Enzimático do Citocromo P-450/biossíntese , Fígado/enzimologia , RNA Mensageiro/biossíntese , Animais , Northern Blotting , Western Blotting , Células Cultivadas , L-Lactato Desidrogenase/metabolismo , Fígado/citologia , Fígado/efeitos dos fármacos , Coelhos
12.
J Lipid Res ; 40(1): 152-9, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9869661

RESUMO

Trans-4-hydroxy-2-nonenal (HNE) is a potent cytotoxic and genotoxic compound originating from the peroxidation of n-6 polyunsaturated fatty acids. Its metabolism has been previously studied in the rat (Alary et al. 1995. Chem. Res. Toxicol., 8: 35-39). In addition to major urinary mercapturic derivatives, some polar urinary metabolites were isolated and could correspond to hydroxylated compounds. 4-Hydroxynonenoic acid (HNA), resulting from the oxidation of the HNE carbonyl group, is a medium chain fatty acid and its omega-hydroxylation might be hypothesized. Therefore, the involvement of the CYP 4A family isoenzymes in the metabolism of [3H]HNE has been investigated in vivo using inducer treatments (fibrates) in wild-type or in peroxisome proliferator-activated receptor alpha (PPARalpha)-deficient mice. In wild-type mice, but not in PPARalpha (-/-) mice, fibrate treatments resulted in an increase of two urinary metabolites characterized, after HPLC purifications and mass spectrometry analyses, as the omega-hydroxylated metabolite of HNA, i.e., 4,9-dihydroxy-2-nonenoic acid, and its oxidized form, 4-hydroxy-2-nonene-1,9-dicarboxylic acid. The formation of the latter is correlated accurately to laurate hydroxylase activity studied concurrently in microsomes prepared from the liver of these animals. Basal levels of these two metabolites were measured in urine of normal and PPARalpha-deficient mice. These results are in accord with an implication of the P450 4A family in the extended oxidative metabolism of 4-HNE.


Assuntos
Aldeídos/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Oxigenases de Função Mista/metabolismo , Receptores Citoplasmáticos e Nucleares/deficiência , Fatores de Transcrição/deficiência , Animais , Cromatografia Líquida de Alta Pressão , Clofibrato/farmacologia , Citocromo P-450 CYP4A , Sistema Enzimático do Citocromo P-450/genética , Feminino , Fenofibrato/farmacologia , Hidroxilação , Hipolipemiantes/farmacologia , Peroxidação de Lipídeos , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcorpos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Oxigenases de Função Mista/genética , Oxirredução , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética
13.
J Biol Chem ; 273(47): 31581-9, 1998 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-9813074

RESUMO

The hepatic CYP4A enzymes are important fatty acid and prostaglandin omega-hydroxylases that are highly inducible by fibric acid hypolipidemic agents and other peroxisome proliferators. Induction of the CYP4A enzymes by peroxisome proliferators is mediated through the nuclear peroxisome proliferator-activated receptor alpha (PPARalpha). Fatty acids have recently been identified as endogenous ligands of PPARalpha, and this receptor has been implicated in the regulation of lipid homeostasis. In the present report we characterized the induction of the hepatic CYP4A genes in rats during the altered lipid metabolism associated with starvation and diabetes. The mRNA levels of CYP4A1, CYP4A2, and CYP4A3 were induced 7-17-fold in the livers of fasted animals and 3-8-fold in the livers of diabetic animals. This was accompanied by corresponding changes in CYP4A protein levels and arachidonic and lauric acid omega-hydroxylase activity. Interestingly, feeding animals after the fasting period caused as much as an 80% suppression of CYP4A mRNA levels, whereas CYP4A protein levels and functional activity returned to control values. A second PPARalpha-responsive gene, acyl-CoA oxidase, was also induced in rat liver by diabetes and fasting. By using PPARalpha-deficient mice, we unambiguously demonstrated that PPARalpha is strictly required for hepatic CYP4A induction by starvation and diabetes. Similarly, induction of hepatic thiolase and bifunctional enzyme also required expression of PPARalpha. This represents the first evidence for the pathophysiologically induced activation of a nuclear receptor.


Assuntos
Adaptação Fisiológica , Sistema Enzimático do Citocromo P-450/biossíntese , Diabetes Mellitus Experimental/metabolismo , Isomerases , Fígado/enzimologia , Oxigenases de Função Mista/biossíntese , Receptores Citoplasmáticos e Nucleares/metabolismo , Inanição/metabolismo , Fatores de Transcrição/metabolismo , 3-Hidroxiacil-CoA Desidrogenases/biossíntese , Acetil-CoA C-Acetiltransferase/biossíntese , Animais , Ácido Araquidônico/metabolismo , Citocromo P-450 CYP4A , Sistema Enzimático do Citocromo P-450/genética , Diabetes Mellitus Experimental/complicações , Enoil-CoA Hidratase/biossíntese , Indução Enzimática , Alimentos , Ácidos Láuricos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microssomos Hepáticos/enzimologia , Oxigenases de Função Mista/genética , Complexos Multienzimáticos/biossíntese , Enzima Bifuncional do Peroxissomo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/genética , Inanição/complicações , Estreptozocina , Fatores de Transcrição/genética
14.
J Biol Chem ; 273(45): 29577-85, 1998 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-9792666

RESUMO

The alpha-isoform of the peroxisome proliferator-activated receptor (PPARalpha) is a nuclear transcription factor activated by structurally diverse chemicals referred to as peroxisome proliferators. Activators can be endogenous molecules (fatty acids/steroids) or xenobiotics (fibrate lipid-lowering drugs). Upon pharmacological activation, PPARalpha modulates target genes encoding lipid metabolism enzymes, lipid transporters, or apolipoproteins, suggesting a role in lipid homeostasis. Transgenic mice deficient in PPARalpha were shown to lack hepatic peroxisomal proliferation and have an impaired expression and induction of several hepatic target genes. Young adult males show hypercholesterolemia but normal triglycerides. Using a long term experimental set up, we identified these mice as a model of monogenic, spontaneous, late onset obesity with stable caloric intake and a marked sexual dimorphism. Serum triglycerides, elevated in aged animals, are higher in females that develop a more pronounced obesity than males. The latter show a marked and original centrilobular-restricted steatosis and a delayed occurrence of obesity. Fat cells from their liver express substantial levels of PPARgamma2 transcripts when compared with lean cells. These studies demonstrate, in rodents, the involvement of PPARalpha nuclear receptor in lipid homeostasis, with a sexually dimorphic control of circulating lipids, fat storage, and obesity. Characterization of this pathological link may help to delineate new molecular targets for therapeutic intervention and could lead to new insights into the etiology and heritability of mammalian obesity.


Assuntos
Fígado Gorduroso/complicações , Hiperlipidemias/genética , Obesidade/complicações , Receptores Citoplasmáticos e Nucleares/genética , Caracteres Sexuais , Fatores de Transcrição/genética , Animais , Fígado Gorduroso/patologia , Feminino , Hiperlipidemias/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
Toxicol Lett ; 98(1-2): 13-23, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9776557

RESUMO

Fibrates modify the expression of genes implicated in lipoprotein and fatty acid metabolism via the peroxisome proliferator-activated receptor alpha(PPARalpha), leading to reductions in serum triglycerides and cholesterol. The expression of certain genes regulated by PPARalpha have been shown to be modified in a species dependent manner. Aspartate aminotransferase (AspAT or GOT) and alanine aminotransferase (AlaAT or GPT) are enzymes involved in intermediate metabolism in all cells and in hepatic gluconeogenesis. These enzymes are also widely used as serum markers of possible tissue damage. This study investigated whether fenofibrate could modify the expression of liver AspAT and/or AlaAT and thus possibly alter transaminase levels independently of a cytotoxic effect. In human Hep G2 cells, fenofibrate increased cytosolic AspAT (cAspAT) activity by 40% and AlaAT activity by 100%, as well as both mRNAs. Nuclear run on assays showed that this effect was, at least in part, transcriptional. Increases in mRNA were also observed in human hepatocyte cultures at concentrations of the drug attained in patients. In C57BL/6 mice, fenofibrate decreased cAspAT and cAlaAT mRNA, while these effects were abolished in PPARalpha knock-out mice. In conclusion, fenofibrate has been shown to modify cAspAT and AlaAT gene expression in a species and PPARalpha dependent manner. This is the first demonstration that cAspAT and AlaAT activities may be pharmacologically altered, independently of a toxic phenomenon.


Assuntos
Fenofibrato/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hipolipemiantes/farmacologia , Proteínas Nucleares/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Transaminases/biossíntese , Transaminases/genética , Fatores de Transcrição/fisiologia , Alanina Transaminase/biossíntese , Alanina Transaminase/genética , Animais , Aspartato Aminotransferases/biossíntese , Aspartato Aminotransferases/genética , Northern Blotting , Células Cultivadas , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Células Tumorais Cultivadas
16.
Arch Biochem Biophys ; 349(2): 349-55, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9448724

RESUMO

In an effort to better understand the role of the 4S benzo(a)pyrene-binding protein in the induction of CYP1A1 by PAHs, we used a genetically engineered mouse line deficient in Ah receptor (AHR -/-). First, we demonstrated through binding experiments analyzed by sucrose gradient sedimentation and gel permeation chromatography that AHR -/- mice have no detectable AHR protein. In contrast, this AHR-deficient line expressed a 4S protein which efficiently binds BP as it does in hepatic cytosol from C57BL/6 mice. In vivo BP exposure in AHR-deficient mice proved the inability to sustain any CYP1A1 mRNA or CYP1A1 protein induction. These findings demonstrate the requirement of an active AHR to sustain the transactivation pathway leading to CYP1A1 induction. Surprisingly, the 4S BP-binding protein, which was previously characterized as the glycine N-methyltransferase, was completely devoid of such an enzymatic activity after purification by Sephacryl gel permeation chromatography. Moreover, sedimentation and chromatographic experiments, under nondenaturing conditions, do not support the assumption of 4S protein as a subunit of a multimeric protein (GNMT) displaying a molecular mass of 150 kDa.


Assuntos
Benzo(a)pireno/farmacologia , Proteínas de Transporte/metabolismo , Citocromo P-450 CYP1A1/biossíntese , Microssomos Hepáticos/metabolismo , Receptores de Hidrocarboneto Arílico/deficiência , Animais , Benzo(a)pireno/metabolismo , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citosol/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microssomos Hepáticos/efeitos dos fármacos , Dibenzodioxinas Policloradas/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/fisiologia , Transativadores
17.
Toxicol Lett ; 102-103: 459-64, 1998 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-10022296

RESUMO

Genetically engineered animal models represent a substantial improvement in in vivo assessment of toxic pathways. Several transgenic mouse lines have been designed to detect specific toxic markers in response to xenobiotic exposure. They are suitable for in vivo large scale screening of potentially toxic effects of drugs and other xenobiotics, and are used as bioassay models for carcinogenicity testing. This contribution will focus on a different strategy, using transgenic knockout mouse lines, to investigate with more accuracy some metabolic pathways leading to the bioactivation or the bioinactivation of xenobiotics. Through direct knockout of cytochrome P450 (CYP) genes or the knockout of the transcriptional activator of CYP genes AHR (aryl hydrocarbon receptor), the involvement of hepatic metabolic enzymes in xenobiotic bioactivation will be exemplified.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/deficiência , Sistema Enzimático do Citocromo P-450/deficiência , Receptores de Hidrocarboneto Arílico/deficiência , Toxicologia , Xenobióticos/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Biotransformação , Sistema Enzimático do Citocromo P-450/genética , Camundongos , Camundongos Knockout , Micotoxinas/farmacocinética , Hidrocarbonetos Policíclicos Aromáticos/metabolismo , Receptores de Hidrocarboneto Arílico/genética
19.
Environ Toxicol Pharmacol ; 2(4): 381-8, 1996 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-21781746

RESUMO

CYP3A is known to be expressed in liver, small intestine and colon. However, its isoform distribution (CYP3A4, 3A5 and 3A7) and inducibility have not been clearly elucidated in the colon. Therefore, we analyzed CYP3A in human colon and compared its expression and inducibility to the human colonic cell lines HT29 and Caco2, which were used as models. Patients suffered either from sigmoiditis or colonic adenocarcinoma. Patients as well as HT29 and Caco2 cells were treated with rifampicin. CYP3A protein expression was analyzed in the colon of patients and in the cells by immunoblot and by isoelectric focusing enabling separation of CYP3A isoforms, while mRNA expression was determined using specific reverse transcription-polymerase chain reaction. In both human colon and cells, CYP3A5 was the main isoform expressed at the protein and mRNA levels. Rifampicin treatment had no effect on CYP3A expression. HT29 and Caco2 cells exhibiting the same CYP3A expression and inducibility might therefore be useful in vitro models for studying xenobiotic metabolism in human colon.

20.
FEBS Lett ; 355(3): 247-50, 1994 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-7988682

RESUMO

HT-29 sublines and Caco-2 clones were analyzed for the expression of cytochrome P-450 3A. The enzyme was found to be expressed in differentiated HT-29 cells selected by resistance to methotrexate and in one of seven Caco-2 clones, TC7. Its expression parallels the differentiation process, with highest levels being observed at late confluency. P-450 3A mRNA and protein patterns, as well as subcellular distribution, are intermediate between those observed in human adult intestine and fetal liver.


Assuntos
Carcinoma/enzimologia , Neoplasias do Colo/enzimologia , Sistema Enzimático do Citocromo P-450/biossíntese , Regulação Neoplásica da Expressão Gênica , Oxigenases de Função Mista/biossíntese , Northern Blotting , Western Blotting , Compartimento Celular , Diferenciação Celular , Células Clonais , Citocromo P-450 CYP2E1 , Sistema Enzimático do Citocromo P-450/genética , Resistência a Medicamentos/genética , Imunofluorescência , Humanos , Metotrexato/farmacologia , Oxigenases de Função Mista/genética , RNA Mensageiro/análise , Seleção Genética , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...