Your browser doesn't support javascript.
loading
Evolutionarily related small viral fusogens hijack distinct but modular actin nucleation pathways to drive cell-cell fusion.
Chan, Ka Man Carmen; Arthur, Ashley L; Morstein, Johannes; Jin, Meiyan; Bhat, Abrar; Schlesinger, Dörte; Son, Sungmin; Stevens, Donté A; Drubin, David G; Fletcher, Daniel A.
Afiliação
  • Chan KMC; University of California Berkeley/University of California San Francisco Graduate Group in Bioengineering, Berkeley, CA 94720.
  • Arthur AL; Department of Bioengineering & Biophysics Group, University of California, Berkeley, CA 94720.
  • Morstein J; Physiology Course, Marine Biological Laboratory, Woods Hole, MA 02543.
  • Jin M; Physiology Course, Marine Biological Laboratory, Woods Hole, MA 02543.
  • Bhat A; Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455.
  • Schlesinger D; Physiology Course, Marine Biological Laboratory, Woods Hole, MA 02543.
  • Son S; Department of Chemistry, New York University, New York, NY 10003.
  • Stevens DA; Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720.
  • Drubin DG; Physiology Course, Marine Biological Laboratory, Woods Hole, MA 02543.
  • Fletcher DA; National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, 560065, India.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article em En | MEDLINE | ID: mdl-33443166
ABSTRACT
Fusion-associated small transmembrane (FAST) proteins are a diverse family of nonstructural viral proteins. Once expressed on the plasma membrane of infected cells, they drive fusion with neighboring cells, increasing viral spread and pathogenicity. Unlike viral fusogens with tall ectodomains that pull two membranes together through conformational changes, FAST proteins have short fusogenic ectodomains that cannot bridge the intermembrane gap between neighboring cells. One orthoreovirus FAST protein, p14, has been shown to hijack the actin cytoskeleton to drive cell-cell fusion, but the actin adaptor-binding motif identified in p14 is not found in any other FAST protein. Here, we report that an evolutionarily divergent FAST protein, p22 from aquareovirus, also hijacks the actin cytoskeleton but does so through different adaptor proteins, Intersectin-1 and Cdc42, that trigger N-WASP-mediated branched actin assembly. We show that despite using different pathways, the cytoplasmic tail of p22 can replace that of p14 to create a potent chimeric fusogen, suggesting they are modular and play similar functional roles. When we directly couple p22 with the parallel filament nucleator formin instead of the branched actin nucleation promoting factor N-WASP, its ability to drive fusion is maintained, suggesting that localized mechanical pressure on the plasma membrane coupled to a membrane-disruptive ectodomain is sufficient to drive cell-cell fusion. This work points to a common biophysical strategy used by FAST proteins to push rather than pull membranes together to drive fusion, one that may be harnessed by other short fusogens responsible for physiological cell-cell fusion.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Actinas / Proteínas de Fusão de Membrana / Fusão de Membrana Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Actinas / Proteínas de Fusão de Membrana / Fusão de Membrana Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article