Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Nutr Biochem ; 22(4): 344-50, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20471816

RESUMO

Ascorbic acid, the active form of vitamin C, is a vital antioxidant in the human liver, yet the molecular mechanisms involved in the regulation of ascorbic acid transporters [human sodium-dependent vitamin C transporters (hSVCT) 1 and 2] in liver cells are poorly understood. Therefore, we characterized the minimal promoter regions of hSVCT1 and 2 in cultured human liver epithelial cells (HepG2) and examined the effects of ascorbic acid deprivation and supplementation on activity and regulation of the transport systems. Identified minimal promoters required for basal activity were found to include multiple cis regulatory elements, whereas mutational analysis demonstrated that HNF-1 sites in the hSVCT1 promoter and KLF/Sp1 sites in the hSVCT2 promoter were essential for activities. When cultured in ascorbic acid deficient or supplemented media, HepG2 cells demonstrated significant (P<.01) and specific reciprocal changes in [(14)C]-Ascorbic acid uptake, and in hSVCT1 mRNA and protein levels as well as hSVCT1 promoter activity. However, no significant changes in hSVCT2 expression or promoter activity were observed during ascorbic acid deficient or supplemented conditions. We mapped the ascorbic acid responsive region in the hSVCT1 promoter and determined that HNF-1 sites are important for the adaptive regulation response. The results of these studies further characterize the hSVCT1 and 2 promoters establish that ascorbic acid uptake by human liver epithelial cells is adaptively regulated and show that transcriptional mechanisms via HNF-1 in the hSVCT1 promoter may, in part, be involved in this regulation.


Assuntos
Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Simportadores/genética , Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Deficiência de Ácido Ascórbico/fisiopatologia , Regulação da Expressão Gênica , Células Hep G2 , Fator 1 Nuclear de Hepatócito/fisiologia , Humanos , Regiões Promotoras Genéticas/fisiologia , Transportadores de Sódio Acoplados à Vitamina C
2.
Neoplasia ; 10(12): 1481-92, 3p following 1492, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19048126

RESUMO

Ovarian carcinoma arises from the ovarian surface epithelium, which undergoes phenotypic changes characteristic of müllerian epithelium during the first stages of tumorigenesis. The variant isoform of the hepatocyte nuclear factor 1 (vHNF1) is a transcription factor involved in the development of tissues derived from the müllerian duct. Here, we show that vHNF1 knockdown in two ovarian carcinoma cell lines, SKOV3 and IGROV1, leads to reduced E-cadherin (E-cadh) expression and decreased proliferation rate. Accordingly, SKOV3 cells ectopically expressing a dominant-negative (DN) vHNF1 mutant undergo an epithelial-mesenchymal-like transition, acquiring a spindle-like morphology, loss of E-cadh, and disrupted cell-cell contacts. Gene expression profiling of DNvHNF1 cells on the basis of a newly compiled list of epithelial-mesenchymal transition-related genes revealed a correlation between vHNF1 loss-of-function and acquisition of the mesenchymal phenotype. Indeed, phenotypic changes were associated with increased Slug transcription and functionality. Accordingly, vHNF1-transfected immortalized ovarian surface epithelial cells showed down-regulation of Snail and Slug transcripts. In DNvHNF1-transfected SKOV3 cells, growth rate decreased, and in vHNF1-transfected immortalized ovarian surface epithelial cells, growth rate increased. By immunohistochemistry, we found a strong association of vHNF1 with E-cadh in clear cell and in a subset of serous carcinomas, data that could potentially contribute in distinguishing different types of ovarian tumors. Our results may help in understanding the biology of ovarian carcinoma, identifying early detection markers, and opening potential avenues for therapeutic intervention.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fator 1 Nuclear de Hepatócito/fisiologia , Neoplasias Ovarianas/metabolismo , Ovário/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Fator 1 Nuclear de Hepatócito/metabolismo , Humanos , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/patologia , Ovário/patologia , Fenótipo , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo
3.
Gene ; 416(1-2): 48-52, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18440731

RESUMO

The human fatty acid binding protein (FABP2) is involved in intestinal absorption and intracellular trafficking of long-chain fatty acids. Here we investigate transcriptional regulation of FABP2 by the endodermal hepatic nuclear factor 1 alpha (HNF-1alpha). In electromobility shift and supershift assays we show the presence of two adjacent HNF-1alpha binding sites within the FABP2 promoter regions -185 to -165 and -169 to -149. HNF-1alpha activates an FABP2 promoter luciferase construct by 3.5 and 20-fold in Caco-2 and Hela cells, respectively. Mutational analysis of HNF-1alpha elements resulted in about 50% reduction of basal and HNF-1alpha induced activity of FABP2 promoter constructs, predominantly caused by deletion of the -185 to -165 site. Thus, our data suggest a major role of HNF-1alpha in control of FABP2 expression in intestine via a functional HNF-1alpha recognition element within FABP2 promoter region -185 to -165.


Assuntos
Proteínas de Ligação a Ácido Graxo/genética , Regulação da Expressão Gênica , Fator 1 Nuclear de Hepatócito/fisiologia , Sequência de Bases , Sítios de Ligação , Células CACO-2 , Células HeLa , Humanos , Mucosa Intestinal/metabolismo , Dados de Sequência Molecular , Mutação , Polimorfismo Genético , Regiões Promotoras Genéticas , Transcrição Gênica , Transfecção
4.
Am J Physiol Endocrinol Metab ; 292(3): E788-801, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17106062

RESUMO

Glucose homeostasis requires the proper expression and regulation of the catalytic subunit of glucose-6-phosphatase (G-6-Pase), which hydrolyzes glucose 6-phosphate to glucose in glucose-producing tissues. Glucose induces the expression of G-6-Pase at the transcriptional and posttranscriptional levels by unknown mechanisms. To better understand this metabolic regulation, we mapped the cis-regulatory elements conferring glucose responsiveness to the rat G-6-Pase gene promoter in glucose-responsive cell lines. The full-length (-4078/+64) promoter conferred a moderate glucose response to a reporter construct in HL1C rat hepatoma cells, which was dependent on coexpression of glucokinase. The same construct provided a robust glucose response in 832/13 INS-1 rat insulinoma cells, which are not glucogenic. Glucose also strongly increased endogenous G-6-Pase mRNA levels in 832/13 cells and in rat pancreatic islets, although the induced levels from islets were still markedly lower than in untreated primary hepatocytes. A distal promoter region was glucose responsive in 832/13 cells and contained a carbohydrate response element with two E-boxes separated by five base pairs. Carbohydrate response element-binding protein bound this region in a glucose-dependent manner in situ. A second, proximal promoter region was glucose responsive in both 832/13 and HL1C cells, with a hepatocyte nuclear factor 1 binding site and two cAMP response elements required for glucose responsiveness. Expression of dominant-negative versions of both cAMP response element-binding protein and CAAT/enhancer-binding protein blocked the glucose response of the proximal region in a dose-dependent manner. We conclude that multiple, distinct cis-regulatory promoter elements are involved in the glucose response of the rat G-6-Pase gene.


Assuntos
Domínio Catalítico , Glucose-6-Fosfatase/genética , Glucose/farmacologia , Regiões Promotoras Genéticas , Elementos de Resposta/efeitos dos fármacos , Animais , Sequência de Bases , Sítios de Ligação , Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Genes Reporter , Fator 1 Nuclear de Hepatócito/fisiologia , Dados de Sequência Molecular , Ratos , Transfecção
5.
Mol Pharmacol ; 70(2): 627-36, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16670373

RESUMO

Hepatitis C virus (HCV) is known to induce hepatic oxidative stress that is implicated in the up-regulation of multidrug resistance proteins (MRPs). The relationship between increased prooxidant production, MRPs, and HCV has not been investigated. Here, we report that a homeodomain-containing transcription factor, hepatocyte nuclear factor (HNF) 1, plays a central role in liver gene regulation during HCV gene expression and/or subgenome replication. MRP2 protein and mRNA expression were increased and MRP2 promoter activity was increased 7-fold. Mutations within the putative HNF1 binding site of the human MRP2 promoter abrogated HCV-induced activation, implicating HNF1 in the induction of MRP2 by HCV. The mechanism by which HNF1-mediated activation occurs seems to be transcriptional, because the regulated expression of HNF4, which is known to control HNF1 expression, was also increased. Consistent with this finding, HNF1 mRNA was increased 10-fold. A promoter-luciferase construct of the human HNF1 gene was activated in an HNF4-dependent manner, and a mutant construct lacking the HNF4 binding site was not activated in HCV-positive cells. Consistent with this hypothesis, HNF4 protein and mRNA levels as well as HNF4 promoter activity and DNA binding activity were increased. The expression of HNF1 seems to play a critical role in the induction of hepatic MRP2 secondary to HCV subgenomic replication. The ability of HCV to induce HNF1 and HNF4 is attributed to 1) increased oxidative stress and 2) direct protein-protein interactions between HCV nonstructural component (NS) 5A and HNF1, leading to enhanced HNF1 DNA binding. In conclusion, we describe a novel mechanism by which HCV gene expression may induce adaptive responses involving MRP2 via HNF1 activation. This may constitute, in part, the cellular detoxification task force during HCV infection.


Assuntos
Regulação da Expressão Gênica , Hepacivirus/patogenicidade , Fator 1 Nuclear de Hepatócito/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Fígado/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Linhagem Celular , DNA/metabolismo , Hepacivirus/genética , Fator 1 Nuclear de Hepatócito/genética , Humanos , Proteína 2 Associada à Farmacorresistência Múltipla , Regiões Promotoras Genéticas , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima , Proteínas não Estruturais Virais/fisiologia , Replicação Viral
6.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 37(1): 35-9, 2006 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-16468637

RESUMO

OBJECTIVE: To investigate the effects of various liver-enriched transcription factors in regulating HBV transcription and replication, and to explore their potential roles in HBV hepatotropism. METHODS: The replication-competent HBV recombinant plasmid pHBV4.1 plus different liver-enriched transcription factor (HNF1, HNF3, HNF4, HNF6, C/EBP and RXRa/PPARa) expression plasmids were co-transfected into nonhepatic cell lines (NIH3T3, HeLa, 293T, SW1353, CV-1 and COS1). The transcription levels of 3.5 kb, 2.4/2.1 kb and 0.7 kb HBV RNA were analyzed by Northern blot hybridization, and the level of HBV DNA replication intermediates was detected by Southern blot hybridization analysis. RESULTS: In the absence of co-transfected liver enriched transcription factor expression vectors, the 3.5 kb HBV RNA is not transcribed and HBV DNA replication is not detected after transfecting of NIH 3T3 cells with pHBV4.1. Expression of the liver-enriched transcription factor HNF4 or RXRalpha/PPARalpha, stimulates the transcription of 3.5 kb HBV RNA and the replication of HBV DNA. In contrast, expression of HNF1, HNF3, HNF6 and C/EBP does not stimulate the transcription of 3.5 kb HBV RNA and therefore does not activate viral replication. HNF4 and RXRalpha/PPARalpha were also shown to activate the transcription of 3.5 kb HBV RNA and viral replication in divers cell types including HeLa, 293T, SW1353, CV-1 and COS1 cells. Mutation of the proximal nucleocapsid HNF4 binding site results in a greatly decreased level of HNF4 or RXRalpha/PPARalpha dependent HBV replication. CONCLUSION: This study demonstrated that the liver-enriched transcription factors HNF4 and RXRa/PPARa can support HBV transcription and replication in nonhepatic cells, indicating that liver-specific gene transcription is one of the determinants of HBV hepatotropism.


Assuntos
Vírus da Hepatite B/genética , Fatores de Transcrição/genética , Transcrição Gênica , Replicação Viral , Animais , Northern Blotting , Southern Blotting , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HeLa , Vírus da Hepatite B/crescimento & desenvolvimento , Fator 1 Nuclear de Hepatócito/genética , Fator 1 Nuclear de Hepatócito/fisiologia , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/fisiologia , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/fisiologia , Fatores Nucleares de Hepatócito/genética , Fatores Nucleares de Hepatócito/fisiologia , Humanos , Camundongos , Células NIH 3T3 , Fatores de Transcrição/fisiologia , Transfecção
7.
Am J Physiol Gastrointest Liver Physiol ; 290(5): G1016-24, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16223943

RESUMO

Hepatocyte nuclear factor-1alpha (HNF-1alpha) is a modified homeodomain-containing transcription factor that has been implicated in the regulation of intestinal genes. To define the importance and underlying mechanism of HNF-1alpha for the regulation of intestinal gene expression in vivo, we analyzed the expression of the intestinal differentiation markers and putative HNF-1alpha targets lactase-phlorizin hydrolase (LPH) and sucrase-isomaltase (SI) in hnf1alpha null mice. We found that in adult jejunum, LPH mRNA in hnf1alpha(-/-) mice was reduced 95% compared with wild-type controls (P < 0.01, n = 4), whereas SI mRNA was virtually identical to that in wild-type mice. Furthermore, SI mRNA abundance was unchanged in the absence of HNF-1alpha along the length of the adult mouse small intestine as well as in newborn jejunum. We found that HNF-1alpha occupies the promoters of both the LPH and SI genes in vivo. However, in contrast to liver and pancreas, where HNF-1alpha regulates target genes by recruitment of histone acetyl transferase activity to the promoter, the histone acetylation state of the LPH and SI promoters was not affected by the presence or absence of HNF-1alpha. Finally, we showed that a subset of hypothesized intestinal target genes is regulated by HNF-1alpha in vivo and that this regulation occurs in a defined tissue-specific and developmental context. These data indicate that HNF-1alpha is an activator of a subset of intestinal genes and induces these genes through an alternative mechanism in which it is dispensable for chromatin remodeling.


Assuntos
Regulação da Expressão Gênica , Fator 1 Nuclear de Hepatócito/metabolismo , Histonas/metabolismo , Lactase-Florizina Hidrolase/genética , Lactase-Florizina Hidrolase/metabolismo , Acetilação , Animais , Genes Reporter , Fator 1 Nuclear de Hepatócito/genética , Fator 1 Nuclear de Hepatócito/fisiologia , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas , Complexo Sacarase-Isomaltase/metabolismo , Fatores de Transcrição/metabolismo
8.
Mech Dev ; 122(12): 1300-9, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16274963

RESUMO

The early transcriptional hierarchy that subdivides the vertebrate hindbrain into seven to eight segments, the rhombomeres (r1-r8), is largely unknown. The Kreisler (MafB, Krml1, Val) gene is earliest gene expressed in an r5/r6-restricted manner and is essential for r5 and r6 development. We have identified the S5 regulatory element that directs early Kreisler expression in the future r5/r6 domain in 0-10 somite stage embryos. variant Hepatocyte Nuclear Factor 1 (vHNF1/HNF1beta/LF-3B) is transiently expressed in the r5/r6 domain of 0-10 somite stage embryos and a vHNF1binding site within this element is essential but not sufficient for r5/r6-specific expression. Thus, early inductive events that initiate Kreisler expression are clearly distinct from later-acting ones that modulate its expression levels. This site and some of the surrounding sequences are evolutionarily conserved in the genomic DNA upstream of the Kreisler gene among species as divergent as mouse, humans, and chickens. This provides the first evidence of a direct requirement for vHNF1 in initiation of Kreisler expression, suggests that the role of vHNF1 is evolutionarily conserved, and indicates that vHNF1 collaborates with other transcription factors, which independently bind to the S5 regulatory region, to establish the r5/r6 domain.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator 1 Nuclear de Hepatócito/fisiologia , Proteínas de Homeodomínio/fisiologia , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/genética , Rombencéfalo/embriologia , Animais , Sequência de Bases , Sítios de Ligação/genética , Diferenciação Celular/genética , Sequência Conservada , Elementos Facilitadores Genéticos , Variação Genética , Fator 1 Nuclear de Hepatócito/biossíntese , Fator 1 Nuclear de Hepatócito/genética , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Rombencéfalo/citologia , Rombencéfalo/metabolismo
9.
Methods Enzymol ; 400: 57-75, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16399343

RESUMO

Glucuronidation, catalyzed by two families of UDP-glucuronosyltransferases (UGTs), represents a major phase II reaction of endo- and xenobiotic biotransformation. UGT1A6 is the founding member of the rat and human UGT1 family. It is expressed in liver and extrahepatic tissues, such as intestine, kidney, testis, and brain, and conjugates planar phenols and arylamines. Serotonin has been identified as a selective endogenous substrate of the human enzyme. UGT1A6 is also involved in conjugation of the drug paracetamol (acetaminophen) and of phenolic metabolites of benzo[a]pyrene (together with rat UGT1A7 and human UGT1A9). High interindividual variability of human liver protein levels is due to a number of influences, including genetic, tissue-specific, and environmental factors. Evidence shows that homo- and heterozygotic expression of UGT1A6 alleles markedly affects enzyme activity. HNF1 may be responsible for tissue-specific UGT1A6 expression. Multiple environmental factors controlling UGT1A6 expression have been identified, including the pregnane X receptor, the constitutive androstane receptor, the aryl hydrocarbon receptor, and Nrf2, a bZIP transcription factor mediating stress responses. However, marked differences have been noted in the expression of rat and human UGT1A6. Regulatory factors have been studied in detail in the human Caco-2 colon adenocarcinoma cell model.


Assuntos
Regulação Enzimológica da Expressão Gênica , Glucuronosiltransferase/genética , Animais , Carcinógenos/metabolismo , Glucuronosiltransferase/classificação , Glucuronosiltransferase/metabolismo , Fator 1 Nuclear de Hepatócito/fisiologia , Humanos , Inativação Metabólica , Ratos , Serotonina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...