Your browser doesn't support javascript.
loading
E2F4's cytoplasmic role in multiciliogenesis is mediated via an N-terminal domain that binds two components of the centriole replication machinery, Deup1 and SAS6.
Hazan, Renin; Mori, Munemasa; Danielian, Paul S; Guen, Vincent J; Rubin, Seth M; Cardoso, Wellington V; Lees, Jacqueline A.
Afiliação
  • Hazan R; David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139.
  • Mori M; Columbia Center for Human Development and Division of Pulmonary, Allergy and Critical Care, Department of Medicine, and.
  • Danielian PS; David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139.
  • Guen VJ; David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139.
  • Rubin SM; Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA 95064.
  • Cardoso WV; Columbia Center for Human Development and Division of Pulmonary, Allergy and Critical Care, Department of Medicine, and.
  • Lees JA; Department of Genetics and Development and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032.
Mol Biol Cell ; 32(20): ar1, 2021 10 01.
Article em En | MEDLINE | ID: mdl-34260288
ABSTRACT
Multiciliated cells play critical roles in the airway, reproductive organs, and brain. Generation of multiple cilia requires both activation of a specialized transcriptional program and subsequent massive amplification of centrioles within the cytoplasm. The E2F4 transcription factor is required for both roles and consequently for multiciliogenesis. Here we establish that E2F4 associates with two distinct components of the centriole replication machinery, Deup1 and SAS6, targeting nonhomologous domains in these proteins. We map Deup1 and SAS6 binding to E2F4's N-terminus and show that this domain is sufficient to mediate E2F4's cytoplasmic role in multiciliogenesis. This sequence is highly conserved across the E2F family, but the ability to bind Deup1 and SAS6 is specific to E2F4 and E2F5, consistent with their shared roles in multiciliogenesis. By generating E2F4/E2F1 chimeras, we identify a six-residue motif that is critical for Deup1 and SAS6 binding. We propose that the ability of E2F4 and E2F5 to recruit Deup1 and/or SAS6, and enable centriole replication, contributes to their cytoplasmic roles in multiciliogenesis.
Assuntos

Texto completo: 1 Coleções: 01-internacional Temas: Geral Base de dados: MEDLINE Assunto principal: Proteínas de Ciclo Celular / Proteínas Supressoras de Tumor / Fator de Transcrição E2F4 / Proteínas Associadas aos Microtúbulos Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Mol Biol Cell Assunto da revista: BIOLOGIA MOLECULAR Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Temas: Geral Base de dados: MEDLINE Assunto principal: Proteínas de Ciclo Celular / Proteínas Supressoras de Tumor / Fator de Transcrição E2F4 / Proteínas Associadas aos Microtúbulos Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Mol Biol Cell Assunto da revista: BIOLOGIA MOLECULAR Ano de publicação: 2021 Tipo de documento: Article