Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 11(1): 5292, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087715

RESUMEN

Recent advances have enabled the direct induction of human tissue-specific stem and progenitor cells from differentiated somatic cells. However, it is not known whether human hepatic progenitor cells (hHepPCs) can be generated from other cell types by direct lineage reprogramming with defined transcription factors. Here, we show that a set of three transcription factors, FOXA3, HNF1A, and HNF6, can induce human umbilical vein endothelial cells to directly acquire the properties of hHepPCs. These induced hHepPCs (hiHepPCs) propagate in long-term monolayer culture and differentiate into functional hepatocytes and cholangiocytes by forming cell aggregates and cystic epithelial spheroids, respectively, under three-dimensional culture conditions. After transplantation, hiHepPC-derived hepatocytes and cholangiocytes reconstitute damaged liver tissues and support hepatic function. The defined transcription factors also induce hiHepPCs from endothelial cells circulating in adult human peripheral blood. These expandable and bipotential hiHepPCs may be useful in the study and treatment of human liver diseases.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Células Endoteliales/citología , Hepatocitos/citología , Células Madre/citología , Animales , Conductos Biliares/citología , Conductos Biliares/fisiología , Agregación Celular , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Células Endoteliales/fisiología , Femenino , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/fisiología , Factor Nuclear 3-gamma del Hepatocito/genética , Factor Nuclear 3-gamma del Hepatocito/fisiología , Factor Nuclear 6 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/fisiología , Hepatocitos/fisiología , Hepatocitos/trasplante , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Esferoides Celulares/citología , Esferoides Celulares/fisiología , Células Madre/fisiología
2.
Am J Physiol Endocrinol Metab ; 314(4): E308-E321, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351489

RESUMEN

The transcription factors pancreatic and duodenal homeobox 1 (Pdx1) and onecut1 (Oc1) are coexpressed in multipotent pancreatic progenitors (MPCs), but their expression patterns diverge in hormone-expressing cells, with Oc1 expression being extinguished in the endocrine lineage and Pdx1 being maintained at high levels in ß-cells. We previously demonstrated that cooperative function of these two factors in MPCs is necessary for proper specification and differentiation of pancreatic endocrine cells. In those studies, we observed a persistent decrease in expression of the ß-cell maturity factor MafA. We therefore hypothesized that Pdx1 and Oc1 cooperativity in MPCs impacts postnatal ß-cell maturation and function. Here our model of Pdx1-Oc1 double heterozygosity was used to investigate the impact of haploinsufficiency for both of these factors on postnatal ß-cell maturation, function, and adaptability. Examining mice at postnatal day (P) 14, we observed alterations in pancreatic insulin content in both Pdx1 heterozygotes and double heterozygotes. Gene expression analysis at this age revealed significantly decreased expression of many genes important for glucose-stimulated insulin secretion (e.g., Glut2, Pcsk1/2, Abcc8) exclusively in double heterozygotes. Analysis of P14 islets revealed an increase in the number of mixed islets in double heterozygotes. We predicted that double-heterozygous ß-cells would have an impaired ability to respond to stress. Indeed, we observed that ß-cell proliferation fails to increase in double heterozygotes in response to either high-fat diet or placental lactogen. We thus report here the importance of cooperation between regulatory factors early in development for postnatal islet maturation and adaptability.


Asunto(s)
Factor Nuclear 6 del Hepatocito/fisiología , Proteínas de Homeodominio/fisiología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/crecimiento & desarrollo , Células Madre Multipotentes/metabolismo , Transactivadores/fisiología , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/genética , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Dieta Alta en Grasa , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Factor Nuclear 6 del Hepatocito/genética , Proteínas de Homeodominio/genética , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/fisiología , Masculino , Ratones , Ratones Transgénicos , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/fisiología , Organogénesis/efectos de los fármacos , Organogénesis/genética , Transactivadores/genética
3.
Biochem Pharmacol ; 93(1): 92-103, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25451687

RESUMEN

Sex-based differences in the pharmacological profiles of many drugs are due in part to the female-predominant expression of CYP3A4, which is the most important CYP isoform responsible for drug metabolism. Transcription factors trigger the sexually dimorphic expression of drug-metabolizing enzymes in response to sex-dependent growth hormone (GH) secretion. We investigated the roles of HNF6, C/EBPα, and RXRα in the regulation of human female-predominant CYP3A4, mouse female-specific CYP3A41, and rat male-specific CYP3A2 expression by GH secretion patterns using HepG2 cells, growth hormone receptor (GHR) knockout mice as well as rat models of orchiectomy and hypophysectomy. The constitutive expression of HNF6 and RXRα was GH-dependent, and GHR deficiency decreased HNF6/C/EBPα complex levels and increased HNF6/RXRα complex levels. Feminine GH secretion induced the binding of HNF6 and C/EBPα to the CYP3A4 and Cyp3a41 promoters and HNF6/C/EBPα complex levels was more efficiently compared with masculine pattern. Additionally, a greater inhibition of the binding of RXRα to the CYP3A4 and Cyp3a41 promoters and HNF6/RXRα complex levels was observed by feminine GH secretion, but less inhibition was observed by masculine pattern. The binding of HNF6, C/EBPα, and RXRα to the CYP3A2 promoter was not directly regulated by androgens. RXRα completely abolished the synergistic activation of the CYP3A4, Cyp3a41, and CYP3A2 promoters by HNF6 and C/EBPα. The results demonstrate that sex-dependent GH secretion patterns affect the expressions and interactions of HNF6, C/EBPα, and RXRα as well as their binding to CYP3A genes. RXRα mediates the sex-dependent influence of GH on CYP3A expression as an important signalling molecule.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/fisiología , Sistema Enzimático del Citocromo P-450/fisiología , Hormona del Crecimiento/metabolismo , Factor Nuclear 6 del Hepatocito/fisiología , Receptor alfa X Retinoide/fisiología , Caracteres Sexuales , Animales , Citocromo P-450 CYP3A , Femenino , Células Hep G2 , Humanos , Hígado/fisiología , Masculino , Ratones , Ratones Noqueados , Ratas , Ratas Wistar
4.
PLoS One ; 9(10): e110194, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25313862

RESUMEN

The entire repertoire of intrinsic factors that control the cell fate determination process of specific retinal neurons has yet to be fully identified. Single cell transcriptome profiling experiments of retinal progenitor cells revealed considerable gene expression heterogeneity between individual cells, especially among different classes of transcription factors. In this study, we show that two of those factors, Onecut1 and Onecut2, are expressed during mouse retinal development. Using mice that are deficient for each of these transcription factors, we further demonstrate a significant loss (∼70-80%) of horizontal cells in the absence of either of these proteins, while the other retinal cells appear at normal numbers. Microarray profiling experiments performed on knockout retinas revealed defects in horizontal cell genes as early as E14.5. Additional profiling assays showed an upregulation of several stress response genes in the adult Onecut2 knockout, suggesting that the integrity of the retina is compromised in the absence of normal numbers of horizontal cells. Interestingly, melanopsin, the gene coding for the photopigment found in photosensitive ganglion cells, was observed to be upregulated in Onecut1 deficient retinas, pointing to a possible regulatory role for Onecut1. Taken together, our data show that similar to Onecut1, Onecut2 is also necessary for the formation of normal numbers of horizontal cells in the developing retina.


Asunto(s)
Factor Nuclear 6 del Hepatocito/fisiología , Proteínas de Homeodominio/fisiología , Retina/crecimiento & desarrollo , Factores de Transcripción/fisiología , Animales , Regulación de la Expresión Génica , Ratones Noqueados , Retina/citología , Retina/metabolismo , Células Horizontales de la Retina/fisiología , Transcriptoma
5.
J Biol Chem ; 288(30): 21648-58, 2013 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-23775071

RESUMEN

The transcription factor MafA is a key regulator of insulin gene expression and maturation of islet ß cells. Despite its importance, the regulatory mechanism of MafA gene expression is still unclear. To identify the transcriptional regulators of MafA, we examined various transcription factors, which are potentially involved in ß cell differentiation. An adenovirus-mediated overexpression study clearly demonstrated that Onecut1 suppresses the promoter activity of MafA through the Foxa2-binding cis-element on the MafA enhancer region (named area A). However, ChIP analysis showed that Foxa2 but not Onecut1 could directly bind to area A. Furthermore, overexpression of Onecut1 inhibited the binding of Foxa2 onto area A upon ChIP analysis. Importantly, insertion of a mutation in the Foxa2-binding site of area A significantly decreased the promoter activity of MafA. These findings suggest that Onecut1 suppresses MafA gene expression through the Foxa2-binding site. In the mouse pancreas, MafA expression was first detected at the latest stage of ß cell differentiation and was scarcely observed in Onecut1-positive cells during pancreas development. In addition, Onecut1 expression was significantly increased in the islets of diabetic db/db mice, whereas MafA expression was markedly decreased. The improved glucose levels of db/db mice with insulin injections significantly reduced Onecut1 expression and rescued the reduction of MafA expression. These in vivo experiments also suggest that Onecut1 is a negative regulator of MafA gene expression. This study implicates the novel role of Onecut1 in the control of normal ß cell differentiation and its involvement in ß cell dysfunction under diabetic conditions by suppressing MafA gene expression.


Asunto(s)
Expresión Génica , Factor Nuclear 6 del Hepatocito/fisiología , Factores de Transcripción Maf de Gran Tamaño/genética , Regiones Promotoras Genéticas/genética , Animales , Sitios de Unión/genética , Western Blotting , Diferenciación Celular/genética , Línea Celular Tumoral , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 6 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/metabolismo , Inmunohistoquímica , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Modelos Genéticos , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Páncreas/metabolismo , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
PLoS One ; 7(12): e50509, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23227180

RESUMEN

The neuromuscular junctions are the specialized synapses whereby spinal motor neurons control the contraction of skeletal muscles. The formation of the neuromuscular junctions is controlled by a complex interplay of multiple mechanisms coordinately activated in motor nerve terminals and in their target myotubes. However, the transcriptional regulators that control in motor neurons the genetic programs involved in neuromuscular junction development remain unknown. Here, we provide evidence that the Onecut transcription factor HNF-6 regulates in motor neurons the formation of the neuromuscular junctions. Indeed, adult Hnf6 mutant mice exhibit hindlimb muscle weakness and abnormal locomotion. This results from defects of hindlimb neuromuscular junctions characterized by an abnormal morphology and defective localization of the synaptic vesicle protein synaptophysin at the motor nerve terminals. These defects are consequences of altered and delayed formation of the neuromuscular junctions in newborn mutant animals. Furthermore, we show that the expression level of numerous regulators of neuromuscular junction formation, namely agrin, neuregulin-2 and TGF-ß receptor II, is downregulated in the spinal motor neurons of Hnf6 mutant newborn animals. Finally, altered formation of neuromuscular junction-like structures in a co-culture model of wildtype myotubes with mutant embryonic spinal cord slices is rescued by recombinant agrin and neuregulin, indicating that depletion in these factors contributes to defective neuromuscular junction development in the absence of HNF-6. Thus, HNF-6 controls in spinal motor neurons a genetic program that coordinates the formation of hindlimb neuromuscular junctions.


Asunto(s)
Factor Nuclear 6 del Hepatocito/fisiología , Neuronas Motoras/fisiología , Unión Neuromuscular/crecimiento & desarrollo , Animales , Secuencia de Bases , Técnicas de Cocultivo , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Locomoción , Ratones , Ratones Mutantes , Microscopía Electrónica , Reacción en Cadena de la Polimerasa
8.
J Comp Neurol ; 520(5): 952-69, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21830221

RESUMEN

Our current study focuses on the expression of two members of the onecut transcription factor family, Onecut1 (Oc1) and Onecut2 (Oc2), in the developing mouse retina. By immunofluorescence staining, we found that Oc1 and Oc2 had very similar expression patterns throughout retinal development. Both factors started to be expressed in the retina at around embryonic day (E) 11.5. At early stages (E11.5 and E12.5), they were expressed in both the neuroblast layer (NBL) and ganglion cell layer (GCL). As development progressed (from E14.5 to postnatal day [P] 0), expression diminished in the retinal progenitor cells and became more restricted to the GCL. By P5, Oc1 and Oc2 were expressed at very low levels in the GCL. By co-labeling with transcription factors known to be involved in retinal ganglion cell (RGC) development, we found that Oc1 and Oc2 had extensive overlap with Math5 in the NBL, and that they completely overlapped with Pou4f2 and Isl1 in the GCL, but only partially in the NBL. Co-labeling of Oc1 with cell cycle markers confirmed that Oc1 was expressed in both proliferating retinal progenitors and postmitotic retinal cells. In addition, we demonstrated that expression of Oc1 and Oc2 did not require Math5, Isl1, or Pou4f2. Thus, Oc1 and Oc2 may regulate the formation of RGCs in a pathway independent of Math5, Pou4f2, and Isl1. Furthermore, we showed that Oc1 and Oc2 were expressed in both developing and mature horizontal cells (HCs). Therefore the two factors may also function in the genesis and maintenance of HCs.


Asunto(s)
Factor Nuclear 6 del Hepatocito/fisiología , Proteínas de Homeodominio/fisiología , Retina/crecimiento & desarrollo , Retina/metabolismo , Factores de Transcripción/fisiología , Animales , Diferenciación Celular/fisiología , Femenino , Factor Nuclear 6 del Hepatocito/deficiencia , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Retina/embriología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Factores de Transcripción/deficiencia
9.
Cell Signal ; 24(1): 9-16, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21893194

RESUMEN

Hepatocyte nuclear factor 6 (HNF6) is one of liver-enriched transcription factors. HNF6 utilizes the bipartite onecut-homeodomain sequence to localize the HNF6 protein to the nuclear compartment and binds to specific DNA sequences of numerous target gene promoters. HNF6 regulates an intricate network and mediates complex biological processes that are best known in the liver and pancreas. The function of HNF6 is correlated to cell proliferation, cell cycle regulation, cell differentiation and organogenesis, cell migration and cell-matrix adhesion, glucose metabolism, bile homeostasis, inflammation and so on. HNF6 controls the transcription of its target genes in different ways. The details of the regulatory pathways and their mechanisms are still under investigation. Future study will explore HNF6 novel functions associated with apoptosis, oncogenesis, and modulation of the inflammatory response. This review highlights recent progression pertaining to the pathophysiologic role of HNF6 and summarizes the potential mechanisms in preclinical animal models. HNF6-mediated pathways represent attractive therapeutic targets for the treatment of the relative diseases such as cholestasis.


Asunto(s)
Factor Nuclear 6 del Hepatocito/fisiología , Animales , Adhesión Celular , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Movimiento Celular , Proliferación Celular , Regulación de la Expresión Génica , Factor Nuclear 6 del Hepatocito/metabolismo , Humanos , Inflamación , Hígado/metabolismo , Hígado/patología , Hepatopatías/metabolismo , Organogénesis/genética , Transcripción Genética
10.
PLoS One ; 5(10): e13344, 2010 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-20967225

RESUMEN

Recently, we demonstrated that the transcription factors HNF6 and FOXA2 function as key regulators in human colorectal liver metastases. To better understand their proposed inhibitory crosstalk, the consequences of functional knockdown of FOXA2 on HNF6 and C/EBPα activity were investigated in the human colon Caco-2 and HepG2 carcinoma cell lines. Specifically, siRNA-mediated gene silencing of FOXA2 repressed transcript expression by >80%. This resulted in a statistically significant 6-, 3-, 4-, and 8-fold increase in mRNA expression of HNF6 and of genes targeted by this transcription factor, e.g., HSP105B, CYP51, and C/EBPα, as determined by qRT-PCR. Thus, functional knockdown of FOXA2 recovered HNF6 activity. Furthermore, with nuclear extracts of Caco-2 cells no HNF6 DNA binding was observed, but expression of HNF1α, FOXA2, FOXA3, and HNF4α protein was abundant. We therefore transfected a plasmid encoding HNF6 into Caco-2 cells but also employed a retroviral vector to transfect HNF6 into HepG2 cells. This resulted in HNF6 protein expression with DNA binding activity being recovered as determined by EMSA band shift assays. Furthermore, by flow cytometry the consequences of HNF6 expression on cell cycle regulation in transfected cells was studied. Essentially, HNF6 inhibited cell cycle progression in the G2/M and G1 phase in Caco-2 and HepG2 cell lines, respectively. Here, proliferation was reduced by 80% and 50% in Caco-2 and HepG2 cells, respectively, as determined by the BrdU labeling assay. Therefore functional knockdown of FOXA2 recovered HNF6 activity and inhibited growth of tumor-cells and may possibly represent a novel therapeutic target in primary and secondary liver malignancies.


Asunto(s)
Carcinoma Hepatocelular/fisiopatología , Neoplasias del Colon/fisiopatología , Factor Nuclear 3-beta del Hepatocito/antagonistas & inhibidores , Factor Nuclear 6 del Hepatocito/fisiología , Neoplasias Hepáticas/fisiopatología , Western Blotting , Carcinoma Hepatocelular/patología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/patología , Ensayo de Cambio de Movilidad Electroforética , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/genética , Humanos , Neoplasias Hepáticas/patología , Interferencia de ARN , ARN Interferente Pequeño , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
11.
Am J Physiol Gastrointest Liver Physiol ; 295(2): G357-66, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18511741

RESUMEN

Growth hormone (GH) function is mediated through multiple endocrine pathways. In the liver, GH also transcriptionally activates hepatocyte nuclear factor-6 (HNF-6; OC-1), a liver-enriched transcription factor that regulates the expression of genes essential to hepatic function. We hypothesize that GH modulates hepatic function in the normal and injured liver through HNF-6 and HNF-6 target genes. CD1 mice received PBS or GH for the 1-, 7-, and 28-day course of Sham operation or bile duct ligation (BDL). Proliferation-, metabolic-, and profibrotic-specific hepatic functions were assessed with a focus on candidate HNF-6 transcriptional target genes. Confirmation of HNF-6 regulation was done by analysis of target gene expression in liver infected with recombinant adenovirus AdHNF-6 expression vectors. GH administration upregulated HNF-6 expression throughout the course of liver injury. This was associated with increased expression of HNF-6 proliferative target genes cyclin D1 and metabolic gene Cyp7A1 and downregulation of profibrogenic TGFb2R. Hepatic function improved such as enhanced hepatocyte proliferation, higher cholesterol clearance throughout the course of injury, and attenuated fibrogenic response at day 28 of BDL. GH treatment also transcriptionally increased albumin expression in an HNF-6-independent manner. This was associated with enhanced serum albumin levels. In conclusion, the GH/HNF-6 axis is a potential in vivo mechanism underlying GH diverse function in the liver to modulate the liver repair response to BDL.


Asunto(s)
Conductos Biliares Intrahepáticos/fisiología , Colestasis Extrahepática/fisiopatología , Factor Nuclear 6 del Hepatocito/fisiología , Hormona de Crecimiento Humana/farmacología , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/biosíntesis , Animales , Proliferación Celular/efectos de los fármacos , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Ciclina D , Ciclinas/biosíntesis , Hepatocitos/citología , Humanos , Ligadura , Regeneración Hepática , Masculino , Ratones , Transportadores de Anión Orgánico Sodio-Dependiente/biosíntesis , Proteínas Recombinantes/farmacología , Simportadores/biosíntesis , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba
12.
Dev Dyn ; 237(1): 124-31, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18095340

RESUMEN

Members of the onecut family of transcription factors play important roles in the development of the liver and pancreas. We have shown previously that onecut1 (hnf6) is important during the terminal stages of intrahepatic biliary development in zebrafish. Here we report the characterization of a third zebrafish onecut gene, onecut3 (oc3), and assay its expression during development and its role in biliary duct formation using morpholino antisense oligonucleotide-mediated knockdown. These experiments reveal an important role for oc3 during the earliest stages of zebrafish biliary development, and suggest that zebrafish oc3 is the functional ortholog of mammalian hnf6, a gene that directs biliary differentiation from bipotential progenitor cells. Consistent with this, zebrafish hnf6 expression was significantly reduced in oc3-deficient larvae. Knockdown of hnf6 in wild-type zebrafish larvae also significantly reduced oc3 expression, suggesting a complex interaction between onecut family member proteins during the latter stages of zebrafish biliary development.


Asunto(s)
Sistema Biliar/embriología , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Secuencia de Aminoácidos , Animales , Sistema Biliar/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Factor Nuclear 6 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/metabolismo , Factor Nuclear 6 del Hepatocito/fisiología , Inmunohistoquímica , Hibridación in Situ , Queratinas/análisis , Larva/metabolismo , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
15.
Hepatology ; 43(2): 276-86, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16440369

RESUMEN

We previously demonstrated that formation of complexes between the DNA-binding domains of hepatocyte nuclear factor 6 (HNF6) and forkhead box a2 (Foxa2) proteins stimulated Foxa2 transcriptional activity. Here, we used HepG2 cell cotransfection assays to demonstrate that HNF6 transcriptional activity was stimulated by CCAAT/enhancer-binding protein alpha (C/EBPalpha), but not by the related C/EBPbeta or C/EBPdelta proteins. Formation of the C/EBPalpha-HNF6 protein complex required the HNF6 cut domain and the C/EBPalpha activation domain (AD) 1/AD2 sequences. This C/EBPalpha-HNF6 transcriptional synergy required both the N-terminal HNF6 polyhistidine and serine/threonine/proline box sequences, as well as the C/EBPalpha AD1/AD2 sequences, the latter of which are known to recruit the CREB binding protein (CBP) transcriptional coactivator. Consistent with these findings, adenovirus E1A-mediated inhibition of p300/CBP histone acetyltransferase activity abrogated C/EBPalpha-HNF6 transcriptional synergy in cotransfection assays. Co-immunoprecipitation assays with liver protein extracts demonstrate an association between the HNF6 and C/EBPalpha transcription factors and the CBP coactivator protein in vivo. Furthermore, chromatin immunoprecipitation assays with hepatoma cells demonstrated that increased levels of both C/EBPalpha and HNF6 proteins were required to stimulate association of these transcription factors and the CBP coactivator protein with the endogenous mouse Foxa2 promoter region. In conclusion, formation of the C/EBPalpha-HNF6 protein complex stimulates recruitment of the CBP coactivator protein for expression of Foxa2, a transcription factor critical for regulating expression of hepatic gluconeogenic genes during fasting.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Proteína de Unión a CREB/metabolismo , Factor Nuclear 6 del Hepatocito/fisiología , Secuencia de Aminoácidos , Animales , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/metabolismo , Humanos , Ratones , Complejos Multiproteicos/fisiología , Regiones Promotoras Genéticas , Transcripción Genética/fisiología , Transfección
16.
Development ; 132(23): 5295-306, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16284120

RESUMEN

Arthrogryposis-renal dysfunction-cholestasis syndrome (ARC) is a rare cause of cholestasis in infants. Causative mutations in VPS33B, a gene that encodes a Class C vacuolar sorting protein, have recently been reported in individuals with ARC. We have identified a zebrafish vps33b-ortholog that is expressed in developing liver and intestine. Knockdown of vps33b causes bile duct paucity and impairs intestinal lipid absorption, thus phenocopying digestive defects characteristic of ARC. By contrast, neither motor axon nor kidney epithelial defects typically seen in ARC could be identified in vps33b-deficient larvae. Biliary defects in vps33b-deficient zebrafish larvae closely resemble the bile duct paucity associated with knockdown of the onecut transcription factor hnf6. Consistent with this, reduced vps33b expression was evident in hnf6-deficient larvae and in larvae with mutation of vhnf1, a downstream target of hnf6. Zebrafish vhnf1, but not hnf6, increases vps33b expression in zebrafish embryos and in mammalian liver cells. Electrophoretic mobility shift assays suggest that this regulation occurs through direct binding of vHnf1 to the vps33b promoter. These findings identify vps33b as a novel downstream target gene of the hnf6/vhnf1 pathway that regulates bile duct development in zebrafish. Furthermore, they show that tissue-specific roles for genes that regulate trafficking of intracellular proteins have been modified during vertebrate evolution.


Asunto(s)
Sistema Biliar/crecimiento & desarrollo , Factor Nuclear 6 del Hepatocito/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Colestasis/etiología , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/metabolismo , Factor Nuclear 6 del Hepatocito/deficiencia , Factor Nuclear 6 del Hepatocito/metabolismo , Humanos , Larva/crecimiento & desarrollo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Mutación , Regiones Promotoras Genéticas , Transporte de Proteínas/genética , Proteínas de Transporte Vesicular , Pez Cebra , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...