Your browser doesn't support javascript.
loading
BAF subunit switching regulates chromatin accessibility to control cell cycle exit in the developing mammalian cortex.
Braun, Simon M G; Petrova, Ralitsa; Tang, Jiong; Krokhotin, Andrey; Miller, Erik L; Tang, Yitai; Panagiotakos, Georgia; Crabtree, Gerald R.
Afiliación
  • Braun SMG; Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA.
  • Petrova R; Department of Developmental Biology, Stanford University, California 94305, USA.
  • Tang J; Department of Pathology, Stanford University, California 94305, USA.
  • Krokhotin A; Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland.
  • Miller EL; Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94143, USA.
  • Tang Y; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143, USA.
  • Panagiotakos G; Kavli Institute for Fundamental Neuroscience, University of California at San Francisco, San Francisco, California 94143, USA.
  • Crabtree GR; Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA.
Genes Dev ; 35(5-6): 335-353, 2021 03 01.
Article en En | MEDLINE | ID: mdl-33602870
ABSTRACT
mSWI/SNF or BAF chromatin regulatory complexes are dosage-sensitive regulators of human neural development frequently mutated in autism spectrum disorders and intellectual disability. Cell cycle exit and differentiation of neural stem/progenitor cells is accompanied by BAF subunit switching to generate neuron-specific nBAF complexes. We manipulated the timing of BAF subunit exchange in vivo and found that early loss of the npBAF subunit BAF53a stalls the cell cycle to disrupt neurogenesis. Loss of BAF53a results in decreased chromatin accessibility at specific neural transcription factor binding sites, including the pioneer factors SOX2 and ASCL1, due to Polycomb accumulation. This results in repression of cell cycle genes, thereby blocking cell cycle progression and differentiation. Cell cycle block upon Baf53a deletion could be rescued by premature expression of the nBAF subunit BAF53b but not by other major drivers of proliferation or differentiation. WNT, EGF, bFGF, SOX2, c-MYC, or PAX6 all fail to maintain proliferation in the absence of BAF53a, highlighting a novel mechanism underlying neural progenitor cell cycle exit in the continued presence of extrinsic proliferative cues.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Cromatina / Proteínas Cromosómicas no Histona / Ciclo Celular / Corteza Cerebelosa / Actinas / Regulación del Desarrollo de la Expresión Génica / Proteínas de Unión al ADN Límite: Animals Idioma: En Revista: Genes Dev Asunto de la revista: BIOLOGIA MOLECULAR Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Cromatina / Proteínas Cromosómicas no Histona / Ciclo Celular / Corteza Cerebelosa / Actinas / Regulación del Desarrollo de la Expresión Génica / Proteínas de Unión al ADN Límite: Animals Idioma: En Revista: Genes Dev Asunto de la revista: BIOLOGIA MOLECULAR Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos