Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Development ; 143(19): 3573-3581, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27578787

RESUMEN

Cell fate specification depends on transcriptional activation driven by lineage-specific transcription factors as well as changes in chromatin organization. To date, the interplay between transcription factors and chromatin modifiers during development is not well understood. We focus here on the initiation of the pancreatic program from multipotent endodermal progenitors. Transcription factors that play key roles in regulating pancreatic progenitor state have been identified, but the chromatin regulators that help to establish and maintain pancreatic fate are less well known. Using a comparative approach, we identify a crucial role for the histone methyltransferase Setd7 in establishing pancreatic cell identity. We show that Setd7 is expressed in the prospective pancreatic endoderm of Xenopus and mouse embryos prior to Pdx1 induction. Importantly, we demonstrate that setd7 is sufficient and required for pancreatic cell fate specification in Xenopus Functional and biochemical approaches in Xenopus and mouse endoderm support that Setd7 modulates methylation marks at pancreatic regulatory regions, possibly through interaction with the transcription factor Foxa2. Together, these results demonstrate that Setd7 acts as a central component of the transcription complex initiating the pancreatic program.


Asunto(s)
Embrión no Mamífero/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Páncreas/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , N-Metiltransferasa de Histona-Lisina/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Inmunoprecipitación , Hibridación in Situ , Ratones , Páncreas/embriología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Xenopus
2.
Semin Cell Dev Biol ; 51: 106-16, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26806634

RESUMEN

Diabetes is a chronic disease caused by the loss or dysfunction of the insulin-producing ß-cells in the pancreas. To date, much of our knowledge about ß-cells in humans comes from studying rare monogenic forms of diabetes. Importantly, the majority of mutations so far associated to monogenic diabetes are in genes that exert a regulatory role in pancreatic development and/or ß-cell function. Thus, the identification and study of novel mutations open an unprecedented window into human pancreatic development. In this review, we summarize major advances in the genetic dissection of different types of monogenic diabetes and the insights gained from a developmental perspective. We highlight future challenges to bridge the gap between the fast accumulation of genetic data through next-generation sequencing and the need of functional insights into disease mechanisms. Lastly, we discuss the relevance and advantages of studying candidate gene variants in vivo using the Xenopus as model system.


Asunto(s)
Diabetes Mellitus/genética , Modelos Animales de Enfermedad , Páncreas/embriología , Xenopus/embriología , Animales , Humanos , Mutación , Páncreas/anomalías , Páncreas/patología , Xenopus/genética , Proteínas de Xenopus/genética
4.
Diabetes ; 63(10): 3557-64, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24848070

RESUMEN

Mutations in several genes cause nonautoimmune diabetes, but numerous patients still have unclear genetic defects, hampering our understanding of the development of the disease and preventing pathogenesis-oriented treatment. We used whole-genome sequencing with linkage analysis to study a consanguineous family with early-onset antibody-negative diabetes and identified a novel deletion in PCBD1 (pterin-4 α-carbinolamine dehydratase/dimerization cofactor of hepatocyte nuclear factor 1 α), a gene that was recently proposed as a likely cause of diabetes. A subsequent reevaluation of patients with mild neonatal hyperphenylalaninemia due to mutations in PCBD1 from the BIODEF database identified three additional patients who had developed HNF1A-like diabetes in puberty, indicating early ß-cell failure. We found that Pcbd1 is expressed in the developing pancreas of both mouse and Xenopus embryos from early specification onward showing colocalization with insulin. Importantly, a morpholino-mediated knockdown in Xenopus revealed that pcbd1 activity is required for the proper establishment of early pancreatic fate within the endoderm. We provide the first genetic evidence that PCBD1 mutations can cause early-onset nonautoimmune diabetes with features similar to dominantly inherited HNF1A-diabetes. This condition responds to and can be treated with oral drugs instead of insulin, which is important clinical information for these patients. Finally, patients at risk can be detected through a newborn screening for phenylketonuria.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Hidroliasas/genética , Mutación , Adolescente , Niño , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Linaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA