Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 10(6): e0132295, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26125584

RESUMEN

Biliary cysts in adult patients affected by polycystic liver disease are lined by cholangiocytes that proliferate, suggesting that initiation of cyst formation depends on proliferation. Here, we challenge this view by analyzing cyst-lining cell proliferation and differentiation in Cpk mouse embryos and in livers from human fetuses affected by Autosomal Recessive Polycystic Kidney Disease (ARPKD), at early stages of cyst formation. Proliferation of fetal cholangiocyte precursors, measured by immunostaining in human and mouse livers, was low and did not differ between normal and ARPKD or Cpk livers, excluding excessive proliferation as an initiating cause of liver cysts. Instead, our analyses provide evidence that the polycystic livers exhibit increased and accelerated differentiation of hepatoblasts into cholangiocyte precursors, eventually coalescing into large biliary cysts. Lineage tracing experiments, performed in mouse embryos, indicated that the cholangiocyte precursors in Cpk mice generate cholangiocytes and periportal hepatocytes, like in wild-type animals. Therefore, contrary to current belief, cyst formation in polycystic liver disease does not necessarily depend on overproliferation. Combining our prenatal data with available data from adult livers, we propose that polycystic liver can be initiated by proliferation-independent mechanisms at a fetal stage, followed by postnatal proliferation-dependent cyst expansion.


Asunto(s)
Sistema Biliar/patología , Proliferación Celular/fisiología , Quiste del Colédoco/patología , Quistes/patología , Hepatopatías/patología , Riñón Poliquístico Autosómico Recesivo/patología , Animales , Enfermedades de los Conductos Biliares/genética , Enfermedades de los Conductos Biliares/patología , Sistema Biliar/citología , Diferenciación Celular , Quistes/genética , Modelos Animales de Enfermedad , Feto/patología , Hepatocitos/citología , Humanos , Hígado/patología , Hepatopatías/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Riñón Poliquístico Autosómico Recesivo/genética , Tamoxifeno/farmacología
2.
J Cell Biol ; 204(5): 821-38, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24567356

RESUMEN

MarvelD3 is a transmembrane component of tight junctions, but there is little evidence for a direct involvement in the junctional permeability barrier. Tight junctions also regulate signaling mechanisms that guide cell proliferation; however, the transmembrane components that link the junction to such signaling pathways are not well understood. In this paper, we show that MarvelD3 is a dynamic junctional regulator of the MEKK1-c-Jun NH2-terminal kinase (JNK) pathway. Loss of MarvelD3 expression in differentiating Caco-2 cells resulted in increased cell migration and proliferation, whereas reexpression in a metastatic tumor cell line inhibited migration, proliferation, and in vivo tumor formation. Expression levels of MarvelD3 inversely correlated with JNK activity, as MarvelD3 recruited MEKK1 to junctions, leading to down-regulation of JNK phosphorylation and inhibition of JNK-regulated transcriptional mechanisms. Interplay between MarvelD3 internalization and JNK activation tuned activation of MEKK1 during osmotic stress, leading to junction dissociation and cell death in MarvelD3-depleted cells. MarvelD3 thus couples tight junctions to the MEKK1-JNK pathway to regulate cell behavior and survival.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/fisiología , Uniones Estrechas/metabolismo , Células CACO-2 , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Humanos , Proteínas de la Membrana/metabolismo , Presión Osmótica
3.
Genome Res ; 23(2): 270-80, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23124521

RESUMEN

In metazoans, the nuclear lamina is thought to play an important role in the spatial organization of interphase chromosomes, by providing anchoring sites for large genomic segments named lamina-associated domains (LADs). Some of these LADs are cell-type specific, while many others appear constitutively associated with the lamina. Constitutive LADs (cLADs) may contribute to a basal chromosome architecture. By comparison of mouse and human lamina interaction maps, we find that the sizes and genomic positions of cLADs are strongly conserved. Moreover, cLADs are depleted of synteny breakpoints, pointing to evolutionary selective pressure to keep cLADs intact. Paradoxically, the overall sequence conservation is low for cLADs. Instead, cLADs are universally characterized by long stretches of DNA of high A/T content. Cell-type specific LADs also tend to adhere to this "A/T rule" in embryonic stem cells, but not in differentiated cells. This suggests that the A/T rule represents a default positioning mechanism that is locally overruled during lineage commitment. Analysis of paralogs suggests that during evolution changes in A/T content have driven the relocation of genes to and from the nuclear lamina, in tight association with changes in expression level. Taken together, these results reveal that the spatial organization of mammalian genomes is highly conserved and tightly linked to local nucleotide composition.


Asunto(s)
Secuencia Rica en At , Secuencia Conservada , Genoma , Lámina Nuclear/metabolismo , Animales , Caenorhabditis elegans , Secuencia Conservada/genética , Drosophila melanogaster , Células Madre Embrionarias/metabolismo , Humanos , Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Ratones , Factor 1 de Transcripción de Unión a Octámeros/metabolismo
4.
J Clin Invest ; 122(5): 1920-32, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22505453

RESUMEN

Prostate cancer (PCa) is a major lethal malignancy in men, but the molecular events and their interplay underlying prostate carcinogenesis remain poorly understood. Epigenetic events and the upregulation of polycomb group silencing proteins including Bmi1 have been described to occur during PCa progression. Here, we found that conditional overexpression of Bmi1 in mice induced prostatic intraepithelial neoplasia, and elicited invasive adenocarcinoma when combined with PTEN haploinsufficiency. In addition, Bmi1 and the PI3K/Akt pathway were coactivated in a substantial fraction of human high-grade tumors. We found that Akt mediated Bmi1 phosphorylation, enhancing its oncogenic potential in an Ink4a/Arf-independent manner. This process also modulated the DNA damage response and affected genomic stability. Together, our findings demonstrate the etiological role of Bmi1 in PCa, unravel an oncogenic collaboration between Bmi1 and the PI3K/Akt pathway, and provide mechanistic insights into the modulation of Bmi1 function by phosphorylation during prostate carcinogenesis.


Asunto(s)
Adenocarcinoma/metabolismo , Reparación del ADN , Proteínas Nucleares/metabolismo , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Roturas del ADN de Doble Cadena , Activación Enzimática , Inestabilidad Genómica , Haploinsuficiencia , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Clasificación del Tumor , Trasplante de Neoplasias , Proteínas Nucleares/genética , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Complejo Represivo Polycomb 1 , Próstata/metabolismo , Próstata/patología , Neoplasia Intraepitelial Prostática/enzimología , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Transducción de Señal , Ubiquitinación
5.
Gastroenterology ; 141(4): 1432-8, 1438.e1-4, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21708104

RESUMEN

UNLABELLED: BACKGROUND& AIMS: Embryonic biliary precursor cells form a periportal sheet called the ductal plate, which is progressively remodeled to generate intrahepatic bile ducts. A limited number of ductal plate cells participate in duct formation; those not involved in duct development are believed to involute by apoptosis. Moreover, cells that express the SRY-related HMG box transcription factor 9 (SOX9), which include the embryonic ductal plate cells, were proposed to continuously supply the liver with hepatic cells. We investigated the role of the ductal plate in hepatic morphogenesis. METHODS: Apoptosis and proliferation were investigated by immunostaining of mouse and human fetal liver tissue. The postnatal progeny of SOX9-expressing ductal plate cells was analyzed after genetic labeling, at the ductal plate stage, by Cre-mediated recombination of a ROSA26RYFP reporter allele. Inducible Cre expression was induced by SOX9 regulatory regions, inserted in a bacterial artificial chromosome. Livers were studied from mice under normal conditions and during diet-induced regeneration. RESULTS: Ductal plate cells did not undergo apoptosis and showed limited proliferation. They generated cholangiocytes lining interlobular bile ducts, bile ductules, and canals of Hering, as well as periportal hepatocytes. Oval cells that appeared during regeneration also derived from the ductal plate. We did not find that liver homeostasis required a continuous supply of cells from SOX9-expressing progenitors. CONCLUSIONS: The ductal plate gives rise to cholangiocytes lining the intrahepatic bile ducts, including its most proximal segments. It also generates periportal hepatocytes and adult hepatic progenitor cells.


Asunto(s)
Células Madre Adultas/fisiología , Conductos Biliares Intrahepáticos/embriología , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/fisiología , Hepatocitos/fisiología , Hígado/embriología , Células Madre Adultas/metabolismo , Animales , Apoptosis , Conductos Biliares Intrahepáticos/metabolismo , Proliferación Celular , Cromosomas Artificiales Bacterianos , Células Madre Embrionarias/metabolismo , Técnica del Anticuerpo Fluorescente , Edad Gestacional , Hepatocitos/metabolismo , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Integrasas/genética , Hígado/metabolismo , Regeneración Hepática , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Ratones , Ratones Transgénicos , Microscopía Confocal , Microscopía Fluorescente , Proteínas/genética , ARN no Traducido , Factor de Transcripción SOX9/biosíntesis , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo
6.
Dev Biol ; 311(2): 579-89, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17936262

RESUMEN

Liver development in mammals is initiated by the formation of a hepatic bud from the ventral foregut endoderm. The hepatic cells then proliferate and invade the septum transversum mesenchyme, and further differentiate to give rise to hepatocytes and biliary cells. By analyzing mice that are knockout for the transcription factors Hepatocyte Nuclear Factor-6 (HNF-6)/Onecut-1 (OC-1) and OC-2, we show here that these factors redundantly stimulate the degradation of the basal lamina surrounding the liver bud and promote hepatoblast migration in the septum transversum. Gene expression analysis indicates that HNF-6 and OC-2 belong to a gene network comprising E-cadherin, thrombospondin-4 and osteopontin, which regulates liver bud expansion by controlling hepatoblast migration and adhesion. This network operating at the onset of liver development contains candidate genes for investigation of liver carcinogenesis.


Asunto(s)
Movimiento Celular/fisiología , Factor Nuclear 6 del Hepatocito/metabolismo , Hepatocitos/fisiología , Proteínas de Homeodominio/metabolismo , Hígado/embriología , Factores de Transcripción/metabolismo , Animales , Adhesión Celular/fisiología , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 6 del Hepatocito/genética , Hepatocitos/citología , Proteínas de Homeodominio/genética , Hígado/anatomía & histología , Hígado/crecimiento & desarrollo , Ratones , Ratones Noqueados , Morfogénesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteopontina/genética , Osteopontina/metabolismo , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
7.
Mol Cell Biol ; 26(16): 6037-46, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16880515

RESUMEN

During liver development, hepatocytes undergo a maturation process that leads to the fully differentiated state. This relies at least in part on the coordinated action of liver-enriched transcription factors (LETFs), but little is known about the dynamics of this coordination. In this context we investigate here the role of the LETF hepatocyte nuclear factor 6 (HNF-6; also called Onecut-1) during hepatocyte differentiation. We show that HNF-6 knockout mouse fetuses have delayed expression of glucose-6-phosphatase (g6pc), which catalyzes the final step of gluconeogenesis and is a late marker of hepatocyte maturation. Using a combination of in vivo and in vitro gain- and loss-of-function approaches, we demonstrate that HNF-6 stimulates endogenous g6pc gene expression directly via a synergistic and interdependent action with HNF-4 and that it involves coordinate recruitment of the coactivator PGC-1alpha. The expression of HNF-6, HNF-4, and PGC-1alpha rises steadily during liver development and precedes that of g6pc. We provide evidence that threshold levels of HNF-6 are required to allow synergism between HNF-6, HNF-4, and PGC-1alpha to induce time-specific expression of g6pc. Our observations on the regulation of g6pc by HNF-6 provide a model whereby synergism, interdependency, and threshold concentrations of LETFs and coactivators determine time-specific expression of genes during liver development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 4 del Hepatocito/metabolismo , Factor Nuclear 6 del Hepatocito/metabolismo , Hígado/embriología , Hígado/metabolismo , Transactivadores/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Células Cultivadas , Embrión de Mamíferos/embriología , Glucosa-6-Fosfatasa/genética , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/deficiencia , Factor Nuclear 6 del Hepatocito/genética , Hepatocitos/citología , Hepatocitos/enzimología , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Células 3T3 NIH , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Transactivadores/genética , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...