Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Appl Toxicol ; 39(12): 1640-1650, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31385618

RESUMEN

Acetaminophen (APAP) is an antipyretic and analgesic, which is commonly associated with drug-induced hepatic injury. C2-ceramide plays a key role in mediating cell life activities, and oltipraz was extensively studied as a cancer chemopreventive agent. Glutathione S-transferase A1 (GSTA1) acts as a vital liver detoxification enzyme. Hepatocyte nuclear factor 1 (HNF-1) regulates various cellular signaling pathways. In this study, we investigated the effects of C2-ceramide and oltipraz on APAP-induced hepatocyte injury and the changes of HNF-1 and GSTA1. Results showed that C2-ceramide (6 µmol/L) exacerbated APAP-induced hepatocyte injury and caused a significant decrease (P < .01) in HNF-1 and GSTA1 expressions. Meanwhile, GSTA1 content in supernatant was significantly increased (P < .01). In contrast, oltipraz (8 µmol/L) reduced the injury and significantly elevated (P < .01) HNF-1 and GSTA1 expressions while GSTA1 content in supernatant was significantly decreased (P < .01). In conclusion, these findings revealed that C2-ceramide inhibited HNF-1 and GSTA1 expression and exacerbated hepatocyte injury, while oltipraz treatment results in the reduction of hepatocyte injury, and promoted HNF-1 and GSTA1 expression. Additionally, the changes in HNF-1 and GSTA1 were related to APAP-induced hepatocyte injury. These results were useful to investigate the mechanism of an antipyretic and analgesic drug combination.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Glutatión Transferasa/metabolismo , Factor Nuclear 1 del Hepatocito/metabolismo , Hepatocitos/efectos de los fármacos , Pirazinas/farmacología , Esfingosina/análogos & derivados , Antioxidantes/metabolismo , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Interacciones Farmacológicas , Expresión Génica/efectos de los fármacos , Glutatión Transferasa/genética , Células Hep G2 , Factor Nuclear 1 del Hepatocito/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Esfingosina/farmacología , Tionas , Tiofenos
2.
J Pharmacol Exp Ther ; 355(3): 429-41, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26427720

RESUMEN

Cytosolic sulfotransferase 1C2 (SULT1C2) is expressed in the kidney, stomach, and liver of rats; however, the mechanisms regulating expression of this enzyme are not known. We evaluated transcriptional regulation of SULT1C2 by mevalonate (MVA)-derived intermediates in primary cultured rat hepatocytes using several cholesterol synthesis inhibitors. Blocking production of mevalonate with the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor pravastatin (30 µM), reduced SULT1C2 mRNA content by ∼40% whereas the squalene synthase inhibitor squalestatin (SQ1, 0.1 µM), which causes accumulation of nonsterol isoprenoids, increased mRNA content by 4-fold. Treatment with MVA (10 mM) strongly induced SULT1C2 mRNA by 12-fold, and this effect was blocked by inhibiting squalene epoxidase but not by more distal cholesterol inhibitors, indicating the effects of MVA are mediated by postsqualene metabolites. Using rapid amplification of cDNA ends (RACE), we characterized the 5' end of SULT1C2 mRNA and used this information to generate constructs for promoter analysis. SQ1 and MVA increased reporter activity by ∼1.6- and 3-fold, respectively, from a construct beginning 49 base pairs (bp) upstream from the longest 5'-RACE product (-3140:-49). Sequence deletions from this construct revealed a hepatocyte nuclear factor 1 (HNF1) element (-2558), and mutation of this element reduced basal (75%) and MVA-induced (30%) reporter activity and attenuated promoter activation following overexpression of HNF1α or 1ß. However, the effects of SQ1 were localized to a more proximal promoter region (-281:-49). Collectively, our findings demonstrate that cholesterol biosynthetic intermediates influence SULT1C2 expression in rat primary hepatocytes. Further, HNF1 appears to play an important role in mediating basal and MVA-induced SULT1C2 transcription.


Asunto(s)
Colesterol/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Hepatocitos/enzimología , Sulfotransferasas/biosíntesis , Sulfotransferasas/genética , Animales , Anticolesterolemiantes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Factor Nuclear 1 del Hepatocito/genética , Factor Nuclear 1 del Hepatocito/metabolismo , Hepatocitos/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Masculino , Ácido Mevalónico/farmacología , Cultivo Primario de Células , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Escualeno-Monooxigenasa/antagonistas & inhibidores , Sulfotransferasas/efectos de los fármacos , Transfección , Ácidos Tricarboxílicos/farmacología
3.
Int J Cancer ; 135(3): 585-97, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24382740

RESUMEN

Targeted approaches have revealed frequent epigenetic alterations in ovarian cancer, but the scope and relation of these changes to histologic subtype of disease is unclear. Genome-wide methylation and expression data for 14 clear cell carcinoma (CCC), 32 non-CCC and four corresponding normal cell lines were generated to determine how methylation profiles differ between cells of different histological derivations of ovarian cancer. Consensus clustering showed that CCC is epigenetically distinct. Inverse relationships between expression and methylation in CCC were identified, suggesting functional regulation by methylation, and included 22 hypomethylated (UM) genes and 276 hypermethylated (HM) genes. Categorical and pathway analyses indicated that the CCC-specific UM genes were involved in response to stress and many contain hepatocyte nuclear factor (HNF) 1-binding sites, while the CCC-specific HM genes included members of the estrogen receptor alpha (ERalpha) network and genes involved in tumor development. We independently validated the methylation status of 17 of these pathway-specific genes, and confirmed increased expression of HNF1 network genes and repression of ERalpha pathway genes in CCC cell lines and primary cancer tissues relative to non-CCC specimens. Treatment of three CCC cell lines with the demethylating agent Decitabine significantly induced expression for all five genes analyzed. Coordinate changes in pathway expression were confirmed using two primary ovarian cancer datasets (p < 0.0001 for both). Our results suggest that methylation regulates specific pathways and biological functions in CCC, with hypomethylation influencing the characteristic biology of the disease while hypermethylation contributes to the carcinogenic process.


Asunto(s)
Adenocarcinoma de Células Claras/genética , Metilación de ADN , Epigenómica , Neoplasias Ováricas/genética , Adenocarcinoma de Células Claras/patología , Femenino , Factor Nuclear 1 del Hepatocito/genética , Factor Nuclear 1 del Hepatocito/metabolismo , Humanos , Estadificación de Neoplasias , Neoplasias Ováricas/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Tumorales Cultivadas
4.
Diabet Med ; 31(6): 721-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24905847

RESUMEN

AIM: To compare the prevalence and clinical features of HNF1ß-related MODY and HNF1α-related MODY in Japanese. METHODS: We enrolled 230 Japanese patients with suspected MODY and examined them for HNF1α and HNF1ß mutations. We characterized the clinical features of HNF1ß-related MODY (HNF1ß-MODY) and HNF1α-related MODY (HNF1α-MODY). RESULTS: Six patients had HNF1ß mutations, four of which were large gene deletions and 24 patients had HNF1α mutations, which included one gene deletion. The mean fasting plasma glucose level at onset of HNF1ß-MODY was considerably higher and the age of onset of HNF1ß-MODY was considerably older than they were for HNF1α-MODY, while the mean BMI and C-peptide index at onset were similar. Three patients with HNF1ß-MODY were found to have dorsal pancreatic agenesis and four of them had whole-gene deletion. Five of the patients with HNF1ß-MODY had insulin secretion defects and were treated with insulin, and four of these did not have a parent with overt diabetes. CONCLUSION: HNF1ß-MODY may present as ß-cell dysfunction in Japanese rather than as hyperinsulinaemia, which it does among European/American. This dysfunction might result from an intrinsically lower capacity for insulin secretion in Japanese. HNF1ß-MODY has an older age of onset than HNF1α-MODY, which may suggest lower penetrance of the disease. In addition, HNF1ß-MODY has a broad spectrum of clinical manifestations, some of which are detectable by imaging. This may be helpful in some cases for selecting HNF1ß-MODY candidates for genetic testing.


Asunto(s)
Pueblo Asiatico/genética , Diabetes Mellitus Tipo 2/genética , Factor Nuclear 1 del Hepatocito/genética , Mutación/genética , Adolescente , Adulto , Edad de Inicio , Análisis de Varianza , Niño , Femenino , Factor Nuclear 1-alfa del Hepatocito/genética , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
5.
J Pharmacol Exp Ther ; 347(1): 181-92, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23922447

RESUMEN

The organic cation transporter 1 (OCT1), also known as solute carrier family 22 member 1, is strongly and specifically expressed in the human liver. Here we show that the hepatocyte nuclear factor 1 (HNF1) regulates OCT1 transcription and contributes to the strong, liver-specific expression of OCT1. Bioinformatic analyses revealed strong conservation of HNF1 binding motifs in an evolutionary conserved region (ECR) in intron 1 of the OCT1 gene. Electrophoretic mobility shift and chromatin immunoprecipitation assays confirmed the specific binding of HNF1 to the intron 1 ECR. In reporter gene assays performed in HepG2 cells, the intron 1 ECR increased SV40 promoter activity by 22-fold and OCT1 promoter activity by 13-fold. The increase was reversed when the HNF1 binding sites in the intron 1 ECR were mutated or the endogenous HNF1α expression was downregulated with small interfering RNA. Following HNF1α overexpression in Huh7 cells, the intron 1 ECR increased SV40 promoter activity by 11-fold and OCT1 promoter activity by 6-fold. Without HNF1α overexpression, the increases were only 3- and 2-fold, respectively. Finally, in human liver samples, high HNF1 expression was significantly correlated with high OCT1 expression (r = 0.48, P = 0.002, n = 40). In conclusion, HNF1 is a strong regulator of OCT1 expression. It remains to be determined whether genetic variants, disease conditions, or drugs that affect HNF1 activity may affect the pharmacokinetics and efficacy of OCT1-transported drugs such as morphine, tropisetron, ondansetron, tramadol, and metformin. Beyond OCT1, this study demonstrates the validity and usefulness of interspecies comparisons in the discovery of functionally relevant genomic sequences.


Asunto(s)
Secuencia Conservada/genética , Evolución Molecular , Factor Nuclear 1 del Hepatocito/genética , Intrones/genética , Transportador 1 de Catión Orgánico/biosíntesis , Transportador 1 de Catión Orgánico/genética , Adolescente , Adulto , Anciano , Animales , Bovinos , Niño , Preescolar , Perros , Femenino , Regulación de la Expresión Génica , Células Hep G2 , Hepatocitos/fisiología , Humanos , Macaca mulatta , Masculino , Ratones , Persona de Mediana Edad , Pan troglodytes , Unión Proteica/genética , Ratas , Especificidad de la Especie , Transcripción Genética , Adulto Joven
6.
J Biol Chem ; 286(8): 6049-60, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21163946

RESUMEN

Resveratrol, a polyphenol compound, is known for its effects on energy homeostasis. With properties of energy sensors mediating effects of calorie restriction, sirtuins are targets of resveratrol. The mammalian sirtuin homolog SIRT1 is a protein deacetylase playing a role in glucose metabolism and islet function. Here, we investigated the effects of resveratrol and possible link with SIRT1 in ß-cells. Insulinoma INS-1E cells and human islets were cultured with resveratrol before analyzing their physiological responses. Treatment of INS-1E cells for 24 h with 25 µM resveratrol resulted in marked potentiation of glucose-stimulated insulin secretion. This effect was associated with elevated glycolytic flux, resulting in increased glucose oxidation, ATP generation, and mitochondrial oxygen consumption. Such changes correlated with up-regulation of key genes for ß-cell function, i.e. Glut2, glucokinase, Pdx-1, Hnf-1α, and Tfam. In human islets, chronic resveratrol treatment similarly increased both the glucose secretory response and expression of the same set of genes, eventually restoring the glucose response in islets obtained from one type 2 diabetic donor. Overexpression of Sirt1 in INS-1E cells potentiated resveratrol effects on insulin secretion. Conversely, inhibition of SIRT1 achieved either by expression of an inactive mutant or by using the EX-527 inhibitor, both abolished resveratrol effects on glucose responses. Treatment of INS-1E cells with EX-527 also prevented resveratrol-induced up-regulation of Glut2, glucokinase, Pdx-1, and Tfam. Resveratrol markedly enhanced the glucose response of INS-1E cells and human islets, even after removal of the compound from the medium. These effects were mediated by and fully dependent on active SIRT1, defining a new role for SIRT1 in the regulation of insulin secretion.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Sirtuina 1/metabolismo , Animales , Carbazoles/farmacología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Glucoquinasa/genética , Glucoquinasa/metabolismo , Glucosa/farmacología , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Factor Nuclear 1 del Hepatocito/genética , Factor Nuclear 1 del Hepatocito/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/citología , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Ratas , Resveratrol , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Estilbenos/farmacología , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
7.
Proc Natl Acad Sci U S A ; 106(47): 19934-9, 2009 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19897727

RESUMEN

The regulated expression of large human genes can depend on long-range interactions to establish appropriate three-dimensional structures across the locus. The cystic fibrosis transmembrane conductance regulator (CFTR) gene, which encompasses 189 kb of genomic DNA, shows a complex pattern of expression with both spatial and temporal regulation. The flanking loci, ASZ1 and CTTNBP2, show very different tissue-specific expression. The mechanisms governing control of CFTR expression remain poorly understood, although they are known to involve intronic regulatory elements. Here, we show a complex looped structure of the CFTR locus in cells that express the gene, which is absent from cells in which the gene is inactive. By using chromatin conformation capture (3C) with a bait probe at the CFTR promoter, we demonstrate close interaction of this region with sequences in the middle of the gene about 100 kb from the promoter and with regions 3' to the locus that are about 200 kb away. We show that these interacting regions correspond to prominent DNase I hypersensitive sites within the locus. Moreover, these sequences act cooperatively in reporter gene constructs and recruit proteins that modify chromatin structure. The model for CFTR gene expression that is revealed by our data provides a paradigm for other large genes with multiple regulatory elements lying within both introns and intergenic regions. We anticipate that these observations will enable original approaches to designing regulated transgenes for tissue-specific gene therapy protocols.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Elementos de Facilitación Genéticos , Células Epiteliales/fisiología , Regulación de la Expresión Génica , Intrones , Conformación de Ácido Nucleico , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/citología , Genes Reporteros , Factor Nuclear 1 del Hepatocito/genética , Factor Nuclear 1 del Hepatocito/metabolismo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
8.
J Microbiol ; 60(10): 1039-1047, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36167927

RESUMEN

Hepatitis B virus (HBV) infection highly increases the risk for liver cirrhosis and hepatocellular carcinoma (HCC). The clinical manifestation of HBV infection is determined by the mutual interplay of the viral genotype, host genetic factors, mode of transmission, adaptive mutations, and environmental factors. Core promoter activation plays a critical role in the pre-genomic RNA transcription of HBV for HBV replication. The mutations of core promoter have been implicated in HCC development. We had obtained HBV genes from Myanmar HBV infectants and identified gene variations at the core promoter region. For measuring the relative transactivation activity on core promoter, we prepared the core-promoter reporter construct. Both of A1762T and G1764A mutation were consistently found in the HBV genes with hepatocellular carcinoma. The A1762T/G1764A mutation was corresponding to K130M/V131I of HBx protein. We prepared the core promoter-luciferase reporter construct containing the double A1762T/G1764A mutation and the K130M/V131I HBx protein expression construct. The A1762T/G1764A mutation highly was responsive to core promoter transactivation by HBx, regardless of HBx mutation. The A1762T/G1764A mutation newly created hepatocyte nuclear factor 1 (HNF1) responsive element. Ectopic expression of HNF1 largely increased the HBV core promoter containing A1762T/G1764A mutation. In addition, hepatic rich fatty acid, palmitic acid and oleic acid, increased K130M/V131I HBx level by core promoter activation. These results provide biological properties and clinical significance of specific HBV core promoter mutants related with HCC development.


Asunto(s)
Carcinoma Hepatocelular , Hepatitis B Crónica , Neoplasias Hepáticas , Carcinoma Hepatocelular/genética , Genotipo , Virus de la Hepatitis B/genética , Hepatitis B Crónica/genética , Factor Nuclear 1 del Hepatocito/genética , Humanos , Neoplasias Hepáticas/genética , Mutación , Nucleótidos , Ácido Oléico , Ácido Palmítico , Regiones Promotoras Genéticas , ARN , Activación Transcripcional
9.
Evol Dev ; 13(1): 38-46, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21210941

RESUMEN

The hindbrain is a vertebrate-specific embryonic structure of the central nervous system formed by iterative transitory units called rhombomeres (r). Rhombomeric cells are segregated by interhombomeric boundaries which are prefigured by sharp gene expression borders. The positioning of the first molecular boundary within the hindbrain (the prospective r4/r5 boundary) responds to the expression of an Iroquois (Irx) gene in the anterior (r4) and the gene vHnf1 at the posterior (r5). However, while Irx3 is expressed anteriorly in amniotes, a novel Irx gene, iro7, acts in teleosts. To assess the evolutionary history of the genes responsible for the positioning of the r4/r5 boundary in vertebrates, we have stepped outside the gnathostomes to investigate these genes in the agnathans Lethenteron japonicum and Petromyzon marinus. We identified one representative of the Hnf1 family in agnathans. Its expression pattern recapitulates that of vHnf1 and Hnf1 in higher vertebrates. Our phylogenetic analysis places this gene basal to gnathostome Hnf1 and vHnf1 genes. We propose that the duplication of an ancestral hnf1 gene present in the common ancestor of agnathans and gnathostomes gave rise to the two genes found in gnathostomes. We have also amplified 3 Irx genes in L. japonicum: LjIrxA, LjIrxC, LjIrxD. The expression pattern of LjIrxA (the agnathan Irx1/3 ortholog) resembles those of Irx3 or iro7 in gnathostomes. We propose that an Irx/hnf1 pair already present in early vertebrates positioned the r4/r5 boundary and that gene duplications occurred in these gene families after the divergence of the agnathans.


Asunto(s)
Evolución Molecular , Proteínas de Peces/genética , Proteínas de Homeodominio/genética , Lampreas/embriología , Lampreas/genética , Rombencéfalo/embriología , Secuencia de Aminoácidos , Animales , Tipificación del Cuerpo , Embrión no Mamífero/embriología , Proteínas de Peces/metabolismo , Duplicación de Gen , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Factor Nuclear 1 del Hepatocito/genética , Lampreas/clasificación , Datos de Secuencia Molecular , Familia de Multigenes , Petromyzon/clasificación , Petromyzon/embriología , Petromyzon/genética , Filogenia , Alineación de Secuencia , Vertebrados/embriología , Vertebrados/genética
10.
Arterioscler Thromb Vasc Biol ; 30(3): 568-74, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20007910

RESUMEN

OBJECTIVE: Our aim was to elucidate mechanisms involved in the acquisition of lipid transport properties during enterocyte differentiation. METHODS AND RESULTS: We show that lipid mobilization via apolipoprotein B lipoproteins is dependent on the expression of microsomal triglyceride transfer protein (MTP) during differentiation of Caco-2 cells into enterocyte-like cells. Mechanistic studies showed that binding of the nuclear receptor family 2 group F member 1 (NR2F1) to the DR1 element in the MTTP promoter suppresses MTTP expression in undifferentiated cells. During cellular differentiation, NR2F1 expression and its binding to MTTP promoter decline and MTP induction ensues. Moreover, undifferentiated cells express inositol-requiring enzyme 1beta (IRE1beta), a protein that posttranscriptionally degrades MTP mRNA, and its expression substantially decreases during differentiation, contributing to MTP induction. Immunohistochemical studies revealed a significant negative relationship between the expressions of MTP and NR2F1/IRE1beta in undifferentiated and differentiated Caco-2 cells, as well as in crypt-villus and jejunum-colon axes of mouse intestine. CONCLUSIONS: We propose that transcriptional and posttranscriptional mechanisms involving NR2F1 and IRE1beta ensure low MTP expression in undifferentiated intestinal cells and avoid apolipoprotein B lipoprotein biosynthesis.


Asunto(s)
Factor de Transcripción COUP I/metabolismo , Proteínas Portadoras/metabolismo , Diferenciación Celular , Endorribonucleasas/metabolismo , Mucosa Intestinal/metabolismo , Lipoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Apolipoproteínas B/metabolismo , Secuencia de Bases , Células CACO-2 , Colon/citología , Colon/metabolismo , Enterocitos/citología , Enterocitos/metabolismo , Factor Nuclear 1 del Hepatocito/genética , Factor Nuclear 1 del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Yeyuno/citología , Yeyuno/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Datos de Secuencia Molecular
11.
BMC Mol Biol ; 11: 9, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-20096102

RESUMEN

BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. We previously demonstrated aberrant methylation of specific CpG dinucleotides in UGT1A1-negative cells, and revealed that methylation state of the UGT1A1 5'-flanking sequence is negatively correlated with gene transcription. Interestingly, one of these CpG dinucleotides (CpG -4) is found close to a HNF1 response element (HRE), known to be involved in activation of UGT1A1 gene expression, and within an upstream stimulating factor (USF) binding site. RESULTS: Gel retardation assays revealed that methylation of CpG-4 directly affect the interaction of USF1/2 with its cognate sequence without altering the binding for HNF1-alpha. Luciferase assays sustained a role for USF1/2 and HNF1-alpha in UGT1A1 regulation in colon cancer cells. Based on the differential expression profiles of HNF1A gene in colon cell lines, we also assessed whether methylation affects its expression. In agreement with the presence of CpG islands in the HNF1A promoter, treatments of UGT1A1-negative HCT116 colon cancer cells with a DNA methyltransferase inhibitor restore HNF1A gene expression, as observed for UGT1A1. CONCLUSIONS: This study reveals that basal UGT1A1 expression in colon cells is positively regulated by HNF1-alpha and USF, and negatively regulated by DNA methylation. Besides, DNA methylation of HNF1A could also play an important role in regulating additional cellular drug metabolism and transporter pathways. This process may contribute to determine local inactivation of drugs such as the anticancer agent SN-38 by glucuronidation and define tumoral response.


Asunto(s)
Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , Glucuronosiltransferasa/genética , Factor Nuclear 1 del Hepatocito/metabolismo , Azacitidina/análogos & derivados , Azacitidina/farmacología , Secuencia de Bases , Neoplasias del Colon/metabolismo , Islas de CpG , Citidina Trifosfato/análogos & derivados , Citidina Trifosfato/farmacología , Metilación de ADN , Ensayo de Cambio de Movilidad Electroforética , Glucuronosiltransferasa/metabolismo , Células HCT116 , Células HT29 , Factor Nuclear 1 del Hepatocito/genética , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Regulación hacia Arriba
12.
Dig Surg ; 27(1): 39-45, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20357450

RESUMEN

Hepatocellular adenomas (HCA) are rare benign tumours occurring mainly in women under oral contraceptives. HCA bleed frequently and transform rarely into hepatocellular carcinoma. Identification of genes recurrently mutated in HCA and good genotype/phenotype correlations provided the basis of a pathomolecular classification of different HCA subgroups, characterized using immunohistochemical markers. HNF1A-mutated HCA: Biallelic-inactivating mutations of HNF1A gene are identified in 35-40% of HCA. HNF1alpha-inactivated HCA display characteristic pathological features, including marked steatosis. The expression of FABP1 (which is a HNF1A target gene) is downregulated and the absence of L-FABP expression diagnosed this subgroup. beta-Catenin-mutated HCA: beta-catenin mutations leading to activation of the Wnt/beta-catenin pathway represented 10-15% of HCA. They are characterized by overexpression of glutamine synthetase and aberrant nuclear beta-catenin staining. These beta-catenin-activated HCA are at greater risk of malignant transformation; they are difficult to differentiate from well-differentiated HCC. Inflammatory HCA (50%): These are defined by the presence of inflammatory infiltrates, sinusoidal dilatation and thick-walled arteries. Small in-frame deletions that target the binding site of gp130 for IL-6 have been reported in 60% of inflammatory HCA. There is an overexpression of the inflammatory proteins serum amyloid A and C-reactive protein in tumour hepatocytes both at mRNA and protein levels. Inflammatory HCA occurred more frequently in patients with high body mass index; they can be also mutated for beta-catenin and therefore are probably at risk of HCC. Unclassified HCA: Less than 10% of HCA do not express any of the above-mentioned phenotypic markers. Taking into account noticeable differences between the HCA subgroups, in terms of clinical and prognostic features, phenotyping may become an important tool for HCA management strategy.


Asunto(s)
Adenoma de Células Hepáticas/clasificación , Neoplasias Hepáticas/clasificación , Adenoma de Células Hepáticas/genética , Adenoma de Células Hepáticas/patología , Biomarcadores de Tumor/análisis , Femenino , Factor Nuclear 1 del Hepatocito/genética , Histocitoquímica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , beta Catenina/genética
13.
Sci Rep ; 10(1): 9999, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32561878

RESUMEN

Brief refeeding times (~60 min) enhanced hepatic Angptl8 expression in fasted mice. We cloned the mouse Angptl8 promoter region to characterise this rapid refeeding-induced increase in hepatic Angptl8 expression. Deletion of the -309/-60 promoter region significantly attenuated basal promoter activity in hepatocytes. A computational motif search revealed a potential binding motif for hepatocyte nuclear factor 1α/1ß (HNF-1α/ß) at -84/-68 bp of the promoter. Mutation of the HNF-1 binding site significantly decreased the promoter activity in hepatocytes, and the promoter carrying the mutated HNF-1 site was not transactivated by co-transfection of HNF-1 in a non-hepatic cell line. Silencing Hnf-1 in hepatoma cells and mouse primary hepatocytes reduced Angptl8 protein levels. Electrophoretic mobility-shift assays confirmed direct binding of Hnf-1 to its Angptl8 promoter binding motif. Hnf-1α expression levels increased after short-term refeeding, paralleling the enhanced in vivo expression of the Angptl8 protein. Chromatin immunoprecipitation (ChIP) confirmed the recruitment of endogenous Hnf-1 to the Angptl8 promoter region. Insulin-treated primary hepatocytes showed increased expression of Angptl8 protein, but knockdown of Hnf-1 completely abolished this enhancement. HNF-1 appears to play essential roles in the rapid refeeding-induced increases in Angptl8 expression. HNF-1α may therefore represent a primary medical target for ANGPTL8-related metabolic abnormalities. The study revealed the transcriptional regulation of the mouse hepatic Angptl8 gene by HNF-1.


Asunto(s)
Proteínas Similares a la Angiopoyetina/genética , Regulación de la Expresión Génica , Factor Nuclear 1 del Hepatocito/genética , Hígado/metabolismo , Transcripción Genética/genética , Proteína 8 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina/metabolismo , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Factor Nuclear 1 del Hepatocito/metabolismo , Hepatocitos/metabolismo , Ratones , Regiones Promotoras Genéticas
14.
Int J Biol Markers ; 35(1): 41-46, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31992114

RESUMEN

OBJECTIVE: α-fetoprotein (AFP) expression is activated during the embryonic stage or hepatocellular carcinogenesis, so it is presumed that AFP is a key endogenous molecule to promote cell proliferation or differentiation. We carried out gene screening in an unknown family with hyper-alpha-fetoproteinemia and some sporadic menopausal women, and discussed the relationship between AFP expression and liver cirrhosis. METHODS: Peripheral blood samples from family members, patients with malignant liver tumors, and normal controls were collected. Full-length sequence of AFP was amplified and directly sequenced, and compared with normal controls. HNF-1α and HNF-1ß in plasma levels of family members, patients with liver cancer, newborns, pregnant women, and normal subjects were detected by ELISA, and the relationship between HNF-1 and AFP mutation or high expression was evaluated. RESULTS: There was a mutation in AFP promoter region at c.-200 C>T, which was located at the binding site of AFP hepatocyte nuclear factor 1 (HNF-1). AFP was higher than 4000 ng/L in all members carrying the mutation, but liver cancer was excluded in the family with hyper-alpha-fetoprotein. However, cirrhosis occurred in post-menopausal women. The cases reviewed showed that unknown hyper-alpha-fetoprotein was closely related to HNF-1 binding point of AFP in post-menopausal women with cirrhosis (7/11), while the plasma levels of HNF-1α and HNF-1ß were not significantly different. CONCLUSION: The mutation of the HNF-1 binding point of AFP may lead to an abnormal high expression of AFP by altering the binding of HNF transcription factors, which is closely related to cirrhosis in menopausal women.


Asunto(s)
Factor Nuclear 1 del Hepatocito/genética , Cirrosis Hepática/genética , Mutación Puntual , alfa-Fetoproteínas/genética , Adulto , Femenino , Factor Nuclear 1 del Hepatocito/sangre , Humanos , Cirrosis Hepática/sangre , Neoplasias Hepáticas/sangre , Neoplasias Hepáticas/genética , Masculino , Persona de Mediana Edad , Linaje , Posmenopausia , Regiones Promotoras Genéticas , Estudios Retrospectivos , alfa-Fetoproteínas/metabolismo
15.
Am J Physiol Cell Physiol ; 297(5): C1220-7, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19741195

RESUMEN

Transport and distribution of vitamin C is primarily regulated by the function of sodium-dependent vitamin C transporters (SVCTs). SVCT1 is expressed in the small intestine, liver, and kidney, organs that play a vital role in whole body vitamin C homeostasis. Despite the importance of this protein, little is known about regulation of the gene encoding SVCT1, Slc23a1. In this study, we present the first investigation of the transcriptional regulation of human Slc23a1, identifying transcription factors that may influence its expression. A 1,239-bp genomic DNA fragment corresponding to the 5'-flanking region of Slc23a1 was isolated from a human hepatocarcinoma cell line (HepG2) and sequenced. When cloned into a reporter gene construct, robust transcriptional activity was seen in this sequence, nearly 25-fold above the control vector. Deletion analysis of the SVCT1 reporter gene vector defined the minimal active promoter as a small 135-bp region upstream of the transcriptional start site. While several transcription factor binding sites were identified within this sequence, reporter constructs showed that basal transcription required the binding of hepatic nuclear factor 1 (HNF-1) to its cognate sequence. Furthermore, mutation of this HNF-1 binding site resulted in complete loss of luciferase expression, even in the context of the whole promoter. Additionally, small interfering RNA knockdown of both members of the HNF-1 family, HNF-1alpha and HNF-1beta, resulted in a significant decline in SVCT1 transcription. Together, these data suggest that HNF-1alpha and/or HNF-1beta binding is required for SVCT1 expression and may be involved in the coordinate regulation of whole body vitamin C status.


Asunto(s)
Regulación de la Expresión Génica , Factor Nuclear 1 del Hepatocito/genética , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Simportadores/genética , Ácido Ascórbico/metabolismo , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Ensayo de Cambio de Movilidad Electroforética , Expresión Génica , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Transportadores de Sodio Acoplados a la Vitamina C , Transcripción Genética
16.
J Surg Res ; 152(2): 209-17, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18701117

RESUMEN

BACKGROUND: The two-layer method (TLM) has recently been found to be superior to simple cold storage in University of Wisconsin (UW) solution as a means of pancreas preservation for islet transplantation. In this study, we investigated whether TLM would result in better hepatocyte function over UW cold storage and if it could be applied to hepatocyte transplantation. MATERIALS AND METHODS: Hepatocytes from male Sprague Dawley rat livers were isolated and divided into three groups: a non-preservation group (group 1), a 10-h preservation group (group 2), and a 24-h preservation group (group 3). Groups 2 and 3 were then divided into three subgroups: a group preserved by the TLM (subgroup a), a group preserved in UW solution (subgroup b), and a group preserved in water (subgroup c). Isolated hepatocytes were evaluated for cell yield, viability, and adenosine triphosphate level after preservation. Hepatocytes were either cultured or transplanted. RESULTS: Although no differences in cell yield or morphological findings were observed between any of the groups, TLM significantly improved hepatocyte viability and adenosine triphosphate levels in comparison with UW cold storage. Albumin production or urea synthesis were significantly higher in subgroup 3a than in subgroup 3b at almost all time points. Surprisingly, after hepatocyte transplantation, the serum albumin level in subgroup 2a was significantly higher than in subgroup 2b at every time point. CONCLUSIONS: The results of this study demonstrated that liver preservation by the TLM before hepatocyte isolation might be beneficial and will be useful in the field of hepatotocyte transplantation.


Asunto(s)
Hepatocitos/citología , Hepatocitos/fisiología , Trasplante de Hígado/métodos , Hígado/fisiología , Preservación de Órganos/métodos , Adenosina , Adenosina Trifosfato/metabolismo , Alopurinol , Animales , Técnicas de Cultivo de Célula , Frío , Glutatión , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Factor Nuclear 1 del Hepatocito/genética , Hepatocitos/trasplante , Insulina , Hígado/citología , Masculino , Soluciones Preservantes de Órganos , ARN/genética , ARN/aislamiento & purificación , Rafinosa , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Albúmina Sérica/biosíntesis , Albúmina Sérica/genética
17.
Mol Pharmacol ; 74(3): 744-54, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18523138

RESUMEN

UDP-glucuronosyltransferases (UGTs) are critical to the detoxification of numerous drugs, environmental pollutants, and endogenous molecules. However, as yet not all of the human UGTs have been cloned and characterized. cDNA clones from the UGT2A3 gene (located on chromosome 4q13) were isolated using pooled human liver RNA. Approximately 10% of clones contained a c.1489A>G nucleotide substitution, yielding proteins with a residue 497 alanine (UGT2A3.2) instead of a threonine (UGT2A3.1). The allele frequency of this polymorphism (rs13128286) was 0.13 in a European-American population as determined by direct DNA sequencing. Of 81 structurally diverse glucuronidation substrates tested, UGT2A3 expressed by a baculovirus system selectively glucuronidated bile acids, particularly hyodeoxycholic acid at the 6-hydroxy position. Apparent K(m) values of UGT2A3.1 and UGT2A3.2 for hyodeoxycholic acid 6-glucuronidation were 69 +/- 7 and 44 +/- 12 microM, respectively. Of 29 different extrahepatic tissues evaluated by real-time polymerase chain reaction, UGT2A3 mRNA was most highly expressed in small intestine (160% of liver), colon (78% of liver), and adipose tissue (91% of liver). An in silico scan of the proximal UGT2A3 promoter/5'-regulatory region identified transcription factor consensus elements consistent with tissue-selective expression in liver (HNF1) and intestine (CXD2), as well as induction by rifampicin (pregnane X receptor). In LS180 human intestinal cells, rifampicin increased UGT2A3 mRNA by more than 4.5-fold compared with vehicle, whereas levels were not significantly affected by the arylhydrocarbon receptor ligand beta-naphthoflavone. This is the first report establishing UGT2A3 as a functional enzyme, and it represents significant progress toward the goal of having a complete set of recombinant human UGTs for comparative functional analyses.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Polimorfismo Genético , Animales , Ácidos y Sales Biliares/metabolismo , Factor de Transcripción CDX2 , Línea Celular , Clonación Molecular , Secuencia de Consenso , Ácido Desoxicólico/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucurónidos/metabolismo , Factor Nuclear 1 del Hepatocito/genética , Proteínas de Homeodominio/genética , Humanos , Cinética , Hígado/enzimología , Especificidad de Órganos/efectos de los fármacos , Receptor X de Pregnano , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Esteroides/genética , Proteínas Recombinantes/metabolismo , Rifampin/farmacología , Activación Transcripcional
18.
Comp Biochem Physiol B Biochem Mol Biol ; 150(4): 358-67, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18514559

RESUMEN

Epinecidin-1, an antimicrobial peptide documented in some fish, is an essential component of the innate immune response in fish, but little is known about its gene regulation. To better understand the molecular mechanism controlling transcription of the epinecidin-1 gene, we cloned and sequenced the genes and promoter regions of three epinecidin-1 peptides from the grouper (Epinephelus coioides). These genes have the potential to encode three putative epinecidin-1 peptides with either a short or a long 5'-untranslated region (UTR). These epinecidin-1 genes, numbered 124-1 (gene structure: 5 exons), 124-2 (gene structure: 5 exons), and 961 (gene structure: 4 exons), have 3' UTR sequences that dramatically differ by being located on different exons in clones 124 and 961. To address the roles of lipopolysaccharide (LPS) and poly(I):poly(C) in regulating epinecidin-1 expression, serial deletions were prepared in the promoter region of two clones that contained three genes. Different fragments of the epinecidin-1 5'-flanking region were transfected into U937 (human histiocytic lymphoma) and ZFL (zebrafish liver) cells and then treated with 0, 1, 10, and 100 mug/mL LPS or poly(I):poly(C). The results showed that after treatment with 10 mug/mL LPS, high promoter activity was observed in the 0.6-kb promoter fragment (of clone 961). Promoter deletions showed that hepatocyte nuclear factor (HNF)-1 was required for a maximal response of epinecidin-1 961 promoter activity after LPS treatment in ZFL cells. Morphological studies of transgenic zebrafish indicated that the 2-kb epinecidin-1 124-1 promoter-driven GFP transcripts appeared in the eye and skin as confirmed by immunohistochemical staining. These results indicate that the 2-kb epinecidin-1 124-1 promoter is active in a tissue-specific manner.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Lubina/genética , Proteínas de Peces/genética , Regiones Promotoras Genéticas/genética , Regiones no Traducidas 3'/genética , Regiones no Traducidas 5'/genética , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Células Cultivadas , Oído , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factor Nuclear 1 del Hepatocito/genética , Factor Nuclear 1 del Hepatocito/metabolismo , Humanos , Inmunohistoquímica , Lipopolisacáridos/farmacología , Hígado/citología , Datos de Secuencia Molecular , Poli I-C/farmacología , Análisis de Secuencia de ADN , Piel/metabolismo , Transfección , Células U937 , Pez Cebra
19.
J Mol Biol ; 364(4): 567-81, 2006 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-17022998

RESUMEN

The orphan nuclear receptor HNF4alpha and the LIM homeodomain factor Isl1 are co-expressed in pancreatic beta-cells and are required for the differentiation and function of these endocrine cells. HNF4alpha activates numerous genes and mutations in its gene are associated with maturity onset diabetes of the young. Cofactors and transcription factors that interact with HNF4alpha are crucial to modulate its transcriptional activity, since the latter is not regulated by conventional ligands. These transcriptional partners interact mainly through the HNF4alpha AF-1 module and the ligand binding domain, which contains the AF-2 module. Here, we showed that Isl1 could enhance the HNF4alpha-mediated activation of transcription of the HNF1alpha, PPARalpha and insulin I promoters. Isl1 interacted with the HNF4alpha AF-2 but also required the HNF4alpha carboxy-terminal F domain for optimal interaction and transcriptional synergy. More specifically, we found that naturally occurring HNF4alpha isoforms, differing only in their F domain, exhibited different abilities to interact and synergize with Isl1, extending the crucial transcriptional modulatory role of the HNF4alpha F domain. HNF4alpha interacted with both the homeodomain and the first LIM domain of Isl1. We found that the transcriptional synergy between HNF4alpha and Isl1 involved an increase in HNF4alpha loading on promoter. The effect was more pronounced on the rat insulin I promoter containing binding sites for both HNF4alpha and Isl1 than on the human HNF1alpha promoter lacking an Isl1 binding site. Moreover, Isl1 could mediate the recruitment of the cofactor CLIM2 resulting in a further transcriptional enhancement of the HNF1alpha promoter activity.


Asunto(s)
Factor Nuclear 4 del Hepatocito/fisiología , Proteínas de Homeodominio/fisiología , Proteínas del Tejido Nervioso/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Animales , Proteínas de Unión al ADN/metabolismo , Factor Nuclear 1 del Hepatocito/genética , Humanos , Insulina/genética , Proteínas con Dominio LIM , Proteínas con Homeodominio LIM , Ligandos , PPAR alfa/genética , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Activación Transcripcional
20.
Mol Syst Biol ; 2: 2006.0029, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16760900

RESUMEN

Although the human genome has been sequenced, progress in understanding gene regulation in humans has been particularly slow. Many computational approaches developed for lower eukaryotes to identify cis-regulatory elements and their associated target genes often do not generalize to mammals, largely due to the degenerate and interactive nature of such elements. Motivated by the switch-like behavior of transcriptional responses, we present a systematic approach that allows adaptive determination of active transcriptional subnetworks (cis-motif combinations, the direct target genes and physiological processes regulated by the corresponding transcription factors) from microarray data in mammals, with accuracy similar to that achieved in lower eukaryotes. Our analysis uncovered several new subnetworks active in human liver and in cell-cycle regulation, with similar functional characteristics as the known ones. We present biochemical evidence for our predictions, and show that the recently discovered G2/M-specific E2F pathway is wider than previously thought; in particular, E2F directly activates certain mitotic genes involved in hepatocellular carcinomas. Additionally, we demonstrate that this method can predict subnetworks in a condition-specific manner, as well as regulatory crosstalk across multiple tissues. Our approach allows systematic understanding of how phenotypic complexity is regulated at the transcription level in mammals and offers marked advantage in systems where little or no prior knowledge of transcriptional regulation is available.


Asunto(s)
Regulación de la Expresión Génica , Modelos Genéticos , Factores de Transcripción/genética , Transcripción Genética , Algoritmos , Animales , Ciclo Celular/genética , Bases de Datos Genéticas , Perfilación de la Expresión Génica/métodos , Genómica , Factor Nuclear 1 del Hepatocito/genética , Factor Nuclear 1 del Hepatocito/metabolismo , Humanos , Hígado/metabolismo , Ratones , Células 3T3 NIH , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA