Your browser doesn't support javascript.
loading
Brg1 Enables Rapid Growth of the Early Embryo by Suppressing Genes That Regulate Apoptosis and Cell Growth Arrest.
Singh, Ajeet P; Foley, Julie F; Rubino, Mark; Boyle, Michael C; Tandon, Arpit; Shah, Ruchir; Archer, Trevor K.
Afiliação
  • Singh AP; Chromatin & Gene Expression Section, Epigenetics & Stem Cell Biology Laboratory, NIEHS, NIH, Research Triangle Park, North Carolina, USA.
  • Foley JF; Special Techniques Group, Cellular & Molecular Pathology Branch, NIEHS, NIH, Research Triangle Park, North Carolina, USA.
  • Rubino M; Chromatin & Gene Expression Section, Epigenetics & Stem Cell Biology Laboratory, NIEHS, NIH, Research Triangle Park, North Carolina, USA.
  • Boyle MC; Chromatin & Gene Expression Section, Epigenetics & Stem Cell Biology Laboratory, NIEHS, NIH, Research Triangle Park, North Carolina, USA.
  • Tandon A; Sciome LLC, Research Triangle Park, North Carolina, USA.
  • Shah R; Sciome LLC, Research Triangle Park, North Carolina, USA.
  • Archer TK; Chromatin & Gene Expression Section, Epigenetics & Stem Cell Biology Laboratory, NIEHS, NIH, Research Triangle Park, North Carolina, USA archer1@niehs.nih.gov.
Mol Cell Biol ; 36(15): 1990-2010, 2016 08 01.
Article em En | MEDLINE | ID: mdl-27185875
ABSTRACT
SWI/SNF (switching/sucrose nonfermenting)-dependent chromatin remodeling establishes coordinated gene expression programs during development, yet important functional details remain to be elucidated. We show that the Brg1 (Brahma-related gene 1; Smarca4) ATPase is globally expressed at high levels during postimplantation development and its conditional ablation, beginning at gastrulation, results in increased apoptosis, growth retardation, and, ultimately, embryonic death. Global gene expression analysis revealed that genes upregulated in Rosa26CreERT2; Brg1(flox/flox) embryos (here referred to as Brg1(d/d) embryos to describe embryos with deletion of the Brg1(flox/flox) alleles) negatively regulate cell cycle progression and cell growth. In addition, the p53 (Trp53) protein, which is virtually undetectable in early wild-type embryos, accumulated in the Brg1(d/d) embryos and activated the p53-dependent pathways. Using P19 cells, we show that Brg1 and CHD4 (chromodomain helicase DNA binding protein 4) coordinate to control target gene expression. Both proteins physically interact and show a substantial overlap of binding sites at chromatin-accessible regions adjacent to genes differentially expressed in the Brg1(d/d) embryos. Specifically, Brg1 deficiency results in reduced levels of the repressive histone H3 lysine K27 trimethylation (H3K27me3) histone mark and an increase in the amount of open chromatin at the regulatory region of the p53 and p21 (Cdkn1a) genes. These results provide insights into the mechanisms by which Brg1 functions, which is in part via the p53 program, to constrain gene expression and facilitate rapid embryonic growth.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Fatores de Transcrição / Proteínas Nucleares / Proteínas / DNA Helicases / Desenvolvimento Embrionário / Pontos de Checagem do Ciclo Celular Limite: Animals Idioma: En Revista: Mol Cell Biol Ano de publicação: 2016 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Fatores de Transcrição / Proteínas Nucleares / Proteínas / DNA Helicases / Desenvolvimento Embrionário / Pontos de Checagem do Ciclo Celular Limite: Animals Idioma: En Revista: Mol Cell Biol Ano de publicação: 2016 Tipo de documento: Article País de afiliação: Estados Unidos