Your browser doesn't support javascript.
loading
Myosin-X is essential to the intercellular spread of HIV-1 Nef through tunneling nanotubes.
Uhl, Jaime; Gujarathi, Shivalee; Waheed, Abdul A; Gordon, Ana; Freed, Eric O; Gousset, Karine.
Afiliação
  • Uhl J; Biology Department, California State University Fresno, Fresno, 93740, USA.
  • Gujarathi S; Biology Department, California State University Fresno, Fresno, 93740, USA.
  • Waheed AA; HIV Dynamics and Replication Program, National Cancer Institute-Frederick, Frederick, MD, 21702, USA.
  • Gordon A; Biology Department, California State University Fresno, Fresno, 93740, USA.
  • Freed EO; HIV Dynamics and Replication Program, National Cancer Institute-Frederick, Frederick, MD, 21702, USA.
  • Gousset K; Biology Department, California State University Fresno, Fresno, 93740, USA. kgousset@csufresno.edu.
J Cell Commun Signal ; 13(2): 209-224, 2019 Jun.
Article em En | MEDLINE | ID: mdl-30443895
ABSTRACT
Tunneling nanotubes (TNTs) are intercellular structures that allow for the passage of vesicles, organelles, genomic material, pathogenic proteins and pathogens. The unconventional actin molecular motor protein Myosin-X (Myo10) is a known inducer of TNTs in neuronal cells, yet its role in other cell types has not been examined. The Nef HIV-1 accessory protein is critical for HIV-1 pathogenesis and can self-disseminate in culture via TNTs. Understanding its intercellular spreading mechanism could reveal ways to control its damaging effects during HIV-1 infection. Our goal in this study was to characterize the intercellular transport mechanism of Nef from macrophages to T cells. We demonstrate that Nef increases TNTs in a Myo10-dependent manner in macrophages and observed the transfer of Nef via TNTs from macrophages to T cells. To quantify this transfer mechanism, we established an indirect flow cytometry assay. Since Nef expression in T cells down-regulates the surface receptor CD4, we correlated the decrease in CD4 to the transfer of Nef between these cells. Thus, we co-cultured macrophages expressing varying levels of Nef with a T cell line expressing high levels of CD4 and quantified the changes in CD4 surface expression resulting from Nef transfer. We demonstrate that Nef transfer occurs via a cell-to-cell dependent mechanism that directly correlates with the presence of Myo10-dependent TNTs. Thus, we show that Nef can regulate Myo10 expression, thereby inducing TNT formation, resulting in its own transfer from macrophages to T cells. In addition, we demonstrate that up-regulation of Myo10 induced by Nef also occurs in human monocyte derived macrophages during HIV-1 infection.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: J Cell Commun Signal Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: J Cell Commun Signal Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Estados Unidos