Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Genes Dev ; 30(21): 2433-2442, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27856615

RESUMEN

Methylated cytosines are associated with gene silencing. The ten-eleven translocation (TET) hydroxylases, which oxidize methylated cytosines to 5-hydroxymethylcytosine (5hmC), are essential for cytosine demethylation. Gene silencing and activation are critical for intestinal stem cell (ISC) maintenance and differentiation, but the potential role of TET hydroxylases in these processes has not yet been examined. Here, we generated genome-wide maps of the 5hmC mark in ISCs and their differentiated progeny. Genes with high levels of hydroxymethylation in ISCs are strongly associated with Wnt signaling and developmental processes. We found Tet1 to be the most abundantly expressed Tet gene in ISCs; therefore, we analyzed intestinal development in Tet1-deficient mice and determined that these mice are growth-retarded, exhibit partial postnatal lethality, and have significantly reduced numbers of proliferative cells in the intestinal epithelium. In addition, the Tet1-deficient intestine displays reduced organoid-forming capacity. In the Tet1-deficient crypt, decreased expression of Wnt target genes such as Axin2 and Lgr5 correlates with lower 5hmC levels at their promoters. These data demonstrate that Tet1-mediated DNA hydroxymethylation plays a critical role in the epigenetic regulation of the Wnt pathway in intestinal stem and progenitor cells and consequently in the self-renewal of the intestinal epithelium.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica/genética , Intestinos/crecimiento & desarrollo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Células Madre/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Intestinos/citología , Ratones , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/genética , Eliminación de Secuencia , Células Madre/citología , Vía de Señalización Wnt/genética
2.
Genes Dev ; 28(6): 652-64, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24637118

RESUMEN

The mammalian intestinal epithelium has a unique organization in which crypts harboring stem cells produce progenitors and finally clonal populations of differentiated cells. Remarkably, the epithelium is replaced every 3-5 d throughout adult life. Disrupted maintenance of the intricate balance of proliferation and differentiation leads to loss of epithelial integrity or barrier function or to cancer. There is a tight correlation between the epigenetic status of genes and expression changes during differentiation; however, the mechanism of how changes in DNA methylation direct gene expression and the progression from stem cells to their differentiated descendants is unclear. Using conditional gene ablation of the maintenance methyltransferase Dnmt1, we demonstrate that reducing DNA methylation causes intestinal crypt expansion in vivo. Determination of the base-resolution DNA methylome in intestinal stem cells and their differentiated descendants shows that DNA methylation is dynamic at enhancers, which are often associated with genes important for both stem cell maintenance and differentiation. We establish that the loss of DNA methylation at intestinal stem cell gene enhancers causes inappropriate gene expression and delayed differentiation.


Asunto(s)
Diferenciación Celular , Metilación de ADN , Intestino Delgado/citología , Células Madre/citología , Animales , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Ratones
3.
Development ; 142(12): 2163-72, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-26023099

RESUMEN

The DNA methyltransferase Dnmt1 maintains DNA methylation patterns and genomic stability in several in vitro cell systems. Ablation of Dnmt1 in mouse embryos causes death at the post-gastrulation stage; however, the functions of Dnmt1 and DNA methylation in organogenesis remain unclear. Here, we report that Dnmt1 is crucial during perinatal intestinal development. Loss of Dnmt1 in intervillus progenitor cells causes global hypomethylation, DNA damage, premature differentiation, apoptosis and, consequently, loss of nascent villi. We further confirm the crucial role of Dnmt1 during crypt development using the in vitro organoid culture system, and illustrate a clear differential requirement for Dnmt1 in immature versus mature organoids. These results demonstrate an essential role for Dnmt1 in maintaining genomic stability during intestinal development and the establishment of intestinal crypts.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/fisiología , Metilación de ADN/genética , Inestabilidad Genómica/genética , Mucosa Intestinal/embriología , Organogénesis/genética , Células Madre/citología , Animales , Apoptosis/genética , Proliferación Celular/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Daño del ADN/genética , Regulación del Desarrollo de la Expresión Génica , Mucosa Intestinal/citología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular
4.
PLoS Genet ; 6(2): e1000848, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20174564

RESUMEN

Transcription factors are key components of regulatory networks that control development, as well as the response to environmental stimuli. We have established an experimental pipeline in Caenorhabditis elegans that permits global identification of the binding sites for transcription factors using chromatin immunoprecipitation and deep sequencing. We describe and validate this strategy, and apply it to the transcription factor PHA-4, which plays critical roles in organ development and other cellular processes. We identified thousands of binding sites for PHA-4 during formation of the embryonic pharynx, and also found a role for this factor during the starvation response. Many binding sites were found to shift dramatically between embryos and starved larvae, from developmentally regulated genes to genes involved in metabolism. These results indicate distinct roles for this regulator in two different biological processes and demonstrate the versatility of transcription factors in mediating diverse biological roles.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/genética , Ambiente , Genoma de los Helmintos/genética , Transactivadores/metabolismo , Animales , Sitios de Unión , Proteínas de Caenorhabditis elegans/genética , Inmunoprecipitación de Cromatina , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes de Helminto/genética , Proteínas Fluorescentes Verdes/metabolismo , Larva/metabolismo , Unión Proteica , ARN Polimerasa II/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Inanición , Análisis de Supervivencia , Transactivadores/genética , Factores de Transcripción/metabolismo
5.
Curr Biol ; 18(18): 1355-64, 2008 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-18804378

RESUMEN

BACKGROUND: FoxA factors are critical regulators of embryonic development and postembryonic life, but little is know about the upstream pathways that modulate their activity. C. elegans pha-4 encodes a FoxA transcription factor that is required to establish the foregut in embryos and to control growth and longevity after birth. We previously identified the AAA+ ATPase homolog ruvb-1 as a potent suppressor of pha-4 mutations. RESULTS: Here we show that ruvb-1 is a component of the Target of Rapamycin (TOR) pathway in C. elegans (CeTOR). Both ruvb-1 and let-363/TOR control nucleolar size and promote localization of box C/D snoRNPs to nucleoli, suggesting a role in rRNA maturation. Inactivation of let-363/TOR or ruvb-1 suppresses the lethality associated with reduced pha-4 activity. The CeTOR pathway controls protein homeostasis and also contributes to adult longevity. We find that pha-4 is required to extend adult lifespan in response to reduced CeTOR signaling. Mutations in the predicted CeTOR target rsks-1/S6 kinase or in ife-2/eIF4E also reduce protein biosynthesis and extend lifespan, but only rsks-1 mutations require pha-4 for adult longevity. In addition, rsks-1, but not ife-2, can suppress the larval lethality associated with pha-4 loss-of-function mutations. CONCLUSIONS: The data suggest that pha-4 and the CeTOR pathway antagonize one another to regulate postembryonic development and adult longevity. We suggest a model in which nutrients promote TOR and S6 kinase signaling, which represses pha-4/FoxA, leading to a shorter lifespan. A similar regulatory hierarchy may function in other animals to modulate metabolism, longevity, or disease.


Asunto(s)
Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Transactivadores/antagonistas & inhibidores , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Larva/fisiología , Longevidad , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/fisiología , Fenotipo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ARN/genética , Interferencia de ARN , Ribonucleoproteínas/fisiología , Transactivadores/genética
6.
Methods Mol Biol ; 1402: 101-117, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26721486

RESUMEN

Chromatin immunoprecipitation with massively parallel DNA sequencing (ChIP-Seq) has been used extensively to determine the genome-wide location of DNA-binding factors, such as transcription factors, posttranscriptionally modified histones, and members of the transcription complex, to assess regulatory input, epigenetic modifications, and transcriptional activity, respectively. Here we describe methods to isolate chromatin from tissues, immunoprecipitate DNA bound to a protein of interest, and perform next-generation sequencing to identify a genome-wide DNA-binding pattern.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Cromatina/aislamiento & purificación , Proteínas de Unión al ADN/aislamiento & purificación , ADN/aislamiento & purificación , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Animales , Cromatina/genética , Cromatina/metabolismo , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Biblioteca de Genes , Genoma , Humanos
7.
Cancer Prev Res (Phila) ; 9(7): 534-46, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26883721

RESUMEN

Intestinal cancer is a heterogeneous disease driven by genetic mutations and epigenetic changes. Approximately 80% of sporadic colorectal cancers are initiated by mutation and inactivation of the adenomatous polyposis coli (APC) gene, which results in unrestrained intestinal epithelial growth and formation of adenomas. Aberrant DNA methylation promotes cancer progression by the inactivation of tumor suppressor genes via promoter methylation. In addition, global DNA hypomethylation is often seen before the formation of adenomas, suggesting that it contributes to neoplastic transformation. Previous studies employed mice with a hypomorphic mutation in DNA methyltransferase 1 (Dnmt1), which exhibited constitutive global DNA hypomethylation and decreased tumorigenesis in the Apc(Min/+) mouse model of intestinal cancer. However, the consequences of intestinal epithelial-specific acute hypomethylation during Apc(Min/+) tumor initiation have not been reported. Using temporally controlled intestinal epithelial-specific gene ablation, we show that total loss of Dnmt1 in the Apc(Min/+) mouse model of intestinal cancer causes accelerated adenoma initiation. Deletion of Dnmt1 precipitates an acute response characterized by hypomethylation of repetitive elements and genomic instability, which surprisingly is followed by remethylation with time. Two months post-Dnmt1 ablation, mice display increased macroadenoma load, consistent with a role for Dnmt1 and DNA methylation in maintaining genomic stability. These data suggest that DNA hypomethylation plays a previously unappreciated role in intestinal adenoma initiation. Cancer Prev Res; 9(7); 534-46. ©2016 AACRSee related article by Lee and Laird, p. 509.


Asunto(s)
Adenoma/genética , Carcinogénesis/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , Neoplasias Intestinales/genética , Adenoma/patología , Animales , ADN (Citosina-5-)-Metiltransferasa 1 , Inestabilidad Genómica/genética , Neoplasias Intestinales/patología , Ratones
8.
Elife ; 52016 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-26808831

RESUMEN

Dnmt1 is critical for immediate postnatal intestinal development, but is not required for the survival of the adult intestinal epithelium, the only rapidly dividing somatic tissue for which this has been shown. Acute Dnmt1 deletion elicits dramatic hypomethylation and genomic instability. Recovery of DNA methylation state and intestinal health is dependent on the de novo methyltransferase Dnmt3b. Ablation of both Dnmt1 and Dnmt3b in the intestinal epithelium is lethal, while deletion of either Dnmt1 or Dnmt3b has no effect on survival. These results demonstrate that Dnmt1 and Dnmt3b cooperate to maintain DNA methylation and genomic integrity in the intestinal epithelium.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/deficiencia , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Mucosa Intestinal/enzimología , Mucosa Intestinal/fisiología , Animales , ADN (Citosina-5-)-Metiltransferasa 1 , Eliminación de Gen , Ratones Noqueados , Análisis de Supervivencia , ADN Metiltransferasa 3B
9.
Artículo en Inglés | MEDLINE | ID: mdl-26236400

RESUMEN

BACKGROUND: DNA methylation has emerged as an important regulator of development and disease, necessitating the design of more efficient and cost-effective methods for detecting and quantifying this epigenetic modification. Next-generation sequencing (NGS) techniques offer single base resolution of CpG methylation levels with high statistical significance, but are also high cost if performed genome-wide. Here, we describe a simplified targeted bisulfite sequencing approach in which DNA sequencing libraries are prepared following sodium bisulfite conversion and two rounds of PCR for target enrichment and sample barcoding, termed BisPCR(2). RESULTS: We have applied the BisPCR(2) technique to validate differential methylation at several type 2 diabetes risk loci identified in genome-wide studies of human islets. We confirmed some previous findings while not others, in addition to identifying novel differentially methylated CpGs at these genes of interest, due to the much higher depth of sequencing coverage in BisPCR(2) compared to prior array-based approaches. CONCLUSION: This study presents a robust, efficient, and cost-effective technique for targeted bisulfite NGS, and illustrates its utility by reanalysis of prior findings from genome-wide studies.

10.
Cold Spring Harb Perspect Biol ; 4(9): a008284, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22952394

RESUMEN

The development of the gastrointestinal tract is a complex process that integrates signaling processes with downstream transcriptional responses. Here, we discuss the regionalization of the primitive gut and formation of the intestine and liver. Anterior-posterior position in the primitive gut is important for establishing regions that will become functional organs. Coordination of signaling between the epithelium and mesenchyme and downstream transcriptional responses is required for intestinal development and homeostasis. Liver development uses a complex transcriptional network that controls the establishment of organ domains, cell differentiation, and adult function. Discussion of these transcriptional mechanisms gives us insight into how the primitive gut, composed of simple endodermal cells, develops into multiple diverse cell types that are organized into complex mature organs.


Asunto(s)
Tipificación del Cuerpo/genética , Redes Reguladoras de Genes , Intestinos/crecimiento & desarrollo , Hígado/crecimiento & desarrollo , Animales , Diferenciación Celular , Desarrollo Embrionario/genética , Homeostasis , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA