Your browser doesn't support javascript.
loading
Myosin 1F Regulates M1-Polarization by Stimulating Intercellular Adhesion in Macrophages.
Piedra-Quintero, Zayda L; Serrano, Carolina; Villegas-Sepúlveda, Nicolás; Maravillas-Montero, José L; Romero-Ramírez, Sandra; Shibayama, Mineko; Medina-Contreras, Oscar; Nava, Porfirio; Santos-Argumedo, Leopoldo.
Afiliação
  • Piedra-Quintero ZL; Department of Molecular Biomedicine, Cinvestav Zacatenco, Mexico City, Mexico.
  • Serrano C; Department of Physiology, Biophysics and Neurosciences, Cinvestav Zacatenco, Mexico City, Mexico.
  • Villegas-Sepúlveda N; Department of Molecular Biomedicine, Cinvestav Zacatenco, Mexico City, Mexico.
  • Maravillas-Montero JL; Research Support Network, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Mexico City, Mexico.
  • Romero-Ramírez S; Research Support Network, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Mexico City, Mexico.
  • Shibayama M; Department of Infectomics and Molecular Pathogenesis, Cinvestav Zacatenco, Mexico City, Mexico.
  • Medina-Contreras O; Immunology and Proteomics Laboratory, Mexico Children's Hospital Federico Gómez, Mexico City, Mexico.
  • Nava P; Department of Physiology, Biophysics and Neurosciences, Cinvestav Zacatenco, Mexico City, Mexico.
  • Santos-Argumedo L; Department of Molecular Biomedicine, Cinvestav Zacatenco, Mexico City, Mexico.
Front Immunol ; 9: 3118, 2018.
Article em En | MEDLINE | ID: mdl-30687322
ABSTRACT
Intestinal macrophages are highly mobile cells with extraordinary plasticity and actively contribute to cytokine-mediated epithelial cell damage. The mechanisms triggering macrophage polarization into a proinflammatory phenotype are unknown. Here, we report that during inflammation macrophages enhance its intercellular adhesion properties in order to acquire a M1-phenotype. Using in vitro and in vivo models we demonstrate that intercellular adhesion is mediated by integrin-αVß3 and relies in the presence of the unconventional class I myosin 1F (Myo1F). Intercellular adhesion mediated by αVß3 stimulates M1-like phenotype in macrophages through hyperactivation of STAT1 and STAT3 downstream of ILK/Akt/mTOR signaling. Inhibition of integrin-αVß3, Akt/mTOR, or lack of Myo1F attenuated the commitment of macrophages into a pro-inflammatory phenotype. In a model of colitis, Myo1F deficiency strongly reduces the secretion of proinflammatory cytokines, decreases epithelial damage, ameliorates disease activity, and enhances tissue repair. Together our findings uncover an unknown role for Myo1F as part of the machinery that regulates intercellular adhesion and polarization in macrophages.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Colite Ulcerativa / Miosina Tipo I / Integrina alfaVbeta3 / Ativação de Macrófagos / Macrófagos Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Colite Ulcerativa / Miosina Tipo I / Integrina alfaVbeta3 / Ativação de Macrófagos / Macrófagos Idioma: En Ano de publicação: 2018 Tipo de documento: Article