Your browser doesn't support javascript.
loading
Myosin IIA interacts with the spectrin-actin membrane skeleton to control red blood cell membrane curvature and deformability.
Smith, Alyson S; Nowak, Roberta B; Zhou, Sitong; Giannetto, Michael; Gokhin, David S; Papoin, Julien; Ghiran, Ionita C; Blanc, Lionel; Wan, Jiandi; Fowler, Velia M.
Afiliação
  • Smith AS; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037.
  • Nowak RB; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037.
  • Zhou S; Microsystems Engineering, Rochester Institute of Technology, Rochester, NY 14623.
  • Giannetto M; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623.
  • Gokhin DS; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14623.
  • Papoin J; Microsystems Engineering, Rochester Institute of Technology, Rochester, NY 14623.
  • Ghiran IC; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14623.
  • Blanc L; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14623.
  • Wan J; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037.
  • Fowler VM; Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY 11030.
Proc Natl Acad Sci U S A ; 115(19): E4377-E4385, 2018 05 08.
Article em En | MEDLINE | ID: mdl-29610350
ABSTRACT
The biconcave disk shape and deformability of mammalian RBCs rely on the membrane skeleton, a viscoelastic network of short, membrane-associated actin filaments (F-actin) cross-linked by long, flexible spectrin tetramers. Nonmuscle myosin II (NMII) motors exert force on diverse F-actin networks to control cell shapes, but a function for NMII contractility in the 2D spectrin-F-actin network of RBCs has not been tested. Here, we show that RBCs contain membrane skeleton-associated NMIIA puncta, identified as bipolar filaments by superresolution fluorescence microscopy. MgATP disrupts NMIIA association with the membrane skeleton, consistent with NMIIA motor domains binding to membrane skeleton F-actin and contributing to membrane mechanical properties. In addition, the phosphorylation of the RBC NMIIA heavy and light chains in vivo indicates active regulation of NMIIA motor activity and filament assembly, while reduced heavy chain phosphorylation of membrane skeleton-associated NMIIA indicates assembly of stable filaments at the membrane. Treatment of RBCs with blebbistatin, an inhibitor of NMII motor activity, decreases the number of NMIIA filaments associated with the membrane and enhances local, nanoscale membrane oscillations, suggesting decreased membrane tension. Blebbistatin-treated RBCs also exhibit elongated shapes, loss of membrane curvature, and enhanced deformability, indicating a role for NMIIA contractility in promoting membrane stiffness and maintaining RBC biconcave disk cell shape. As structures similar to the RBC membrane skeleton exist in many metazoan cell types, these data demonstrate a general function for NMII in controlling specialized membrane morphology and mechanical properties through contractile interactions with short F-actin in spectrin-F-actin networks.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Actinas / Miosina não Muscular Tipo IIA / Forma Celular / Membrana Eritrocítica Limite: Humans Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Actinas / Miosina não Muscular Tipo IIA / Forma Celular / Membrana Eritrocítica Limite: Humans Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2018 Tipo de documento: Article