Your browser doesn't support javascript.
loading
Endosomal egress and intercellular transmission of hepatic ApoE-containing lipoproteins and its exploitation by the hepatitis C virus.
Pham, Minh-Tu; Lee, Ji-Young; Ritter, Christian; Thielemann, Roman; Meyer, Janis; Haselmann, Uta; Funaya, Charlotta; Laketa, Vibor; Rohr, Karl; Bartenschlager, Ralf.
Afiliação
  • Pham MT; Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany.
  • Lee JY; German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.
  • Ritter C; Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany.
  • Thielemann R; German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.
  • Meyer J; BioQuant Center, IPMB, Biomedical Computer Vision Group, Heidelberg University, Heidelberg, Germany.
  • Haselmann U; BioQuant Center, IPMB, Biomedical Computer Vision Group, Heidelberg University, Heidelberg, Germany.
  • Funaya C; BioQuant Center, IPMB, Biomedical Computer Vision Group, Heidelberg University, Heidelberg, Germany.
  • Laketa V; Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany.
  • Rohr K; Electron Microscopy Core Facility (EMCF), Heidelberg University, Heidelberg, Germany.
  • Bartenschlager R; German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.
PLoS Pathog ; 19(7): e1011052, 2023 07.
Article em En | MEDLINE | ID: mdl-37506130
ABSTRACT
Liver-generated plasma Apolipoprotein E (ApoE)-containing lipoproteins (LPs) (ApoE-LPs) play central roles in lipid transport and metabolism. Perturbations of ApoE can result in several metabolic disorders and ApoE genotypes have been associated with multiple diseases. ApoE is synthesized at the endoplasmic reticulum and transported to the Golgi apparatus for LP assembly; however, the ApoE-LPs transport pathway from there to the plasma membrane is largely unknown. Here, we established an integrative imaging approach based on a fully functional fluorescently tagged ApoE. We found that newly synthesized ApoE-LPs accumulate in CD63-positive endosomes of hepatocytes. In addition, we observed the co-egress of ApoE-LPs and CD63-positive intraluminal vesicles (ILVs), which are precursors of extracellular vesicles (EVs), along the late endosomal trafficking route in a microtubule-dependent manner. A fraction of ApoE-LPs associated with CD63-positive EVs appears to be co-transmitted from cell to cell. Given the important role of ApoE in viral infections, we employed as well-studied model the hepatitis C virus (HCV) and found that the viral replicase component nonstructural protein 5A (NS5A) is enriched in ApoE-containing ILVs. Interaction between NS5A and ApoE is required for the efficient release of ILVs containing HCV RNA. These vesicles are transported along the endosomal ApoE egress pathway. Taken together, our data argue for endosomal egress and transmission of hepatic ApoE-LPs, a pathway that is hijacked by HCV. Given the more general role of EV-mediated cell-to-cell communication, these insights provide new starting points for research into the pathophysiology of ApoE-related metabolic and infection-related disorders.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Hepatite C / Hepacivirus Limite: Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Hepatite C / Hepacivirus Limite: Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article