Endosomal egress and intercellular transmission of hepatic ApoE-containing lipoproteins and its exploitation by the hepatitis C virus

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

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.

In: PLoS Pathogens, Vol. 19, No. 7, e1011052, 2023.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Pham, M-T, Lee, J-Y, Ritter, C, Thielemann, R, Meyer, J, Haselmann, U, Funaya, C, Laketa, V, Rohr, K & Bartenschlager, R 2023, 'Endosomal egress and intercellular transmission of hepatic ApoE-containing lipoproteins and its exploitation by the hepatitis C virus', PLoS Pathogens, vol. 19, no. 7, e1011052. https://doi.org/10.1371/journal.ppat.1011052

APA

Pham, M-T., Lee, J-Y., Ritter, C., Thielemann, R., Meyer, J., Haselmann, U., Funaya, C., Laketa, V., Rohr, K., & Bartenschlager, R. (2023). Endosomal egress and intercellular transmission of hepatic ApoE-containing lipoproteins and its exploitation by the hepatitis C virus. PLoS Pathogens, 19(7), [e1011052]. https://doi.org/10.1371/journal.ppat.1011052

Vancouver

Pham M-T, Lee J-Y, Ritter C, Thielemann R, Meyer J, Haselmann U et al. Endosomal egress and intercellular transmission of hepatic ApoE-containing lipoproteins and its exploitation by the hepatitis C virus. PLoS Pathogens. 2023;19(7). e1011052. https://doi.org/10.1371/journal.ppat.1011052

Author

Pham, Minh-Tu ; Lee, Ji-Young ; Ritter, Christian ; Thielemann, Roman ; Meyer, Janis ; Haselmann, Uta ; Funaya, Charlotta ; Laketa, Vibor ; Rohr, Karl ; Bartenschlager, Ralf. / Endosomal egress and intercellular transmission of hepatic ApoE-containing lipoproteins and its exploitation by the hepatitis C virus. In: PLoS Pathogens. 2023 ; Vol. 19, No. 7.

Bibtex

@article{913262faad3642a2b4556a223016ead3,
title = "Endosomal egress and intercellular transmission of hepatic ApoE-containing lipoproteins and its exploitation by the hepatitis C virus",
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.",
keywords = "Humans, Hepacivirus/physiology, Lipopolysaccharides/metabolism, Virus Assembly/physiology, Hepatitis C, Endosomes/metabolism, Apolipoproteins E/metabolism",
author = "Minh-Tu Pham and Ji-Young Lee and Christian Ritter and Roman Thielemann and Janis Meyer and Uta Haselmann and Charlotta Funaya and Vibor Laketa and Karl Rohr and Ralf Bartenschlager",
note = "Copyright: {\textcopyright} 2023 Pham et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.",
year = "2023",
doi = "10.1371/journal.ppat.1011052",
language = "English",
volume = "19",
journal = "P L o S Pathogens (Online)",
issn = "1553-7374",
publisher = "public library of science",
number = "7",

}

RIS

TY - JOUR

T1 - Endosomal egress and intercellular transmission of hepatic ApoE-containing lipoproteins and its exploitation by the hepatitis C virus

AU - Pham, Minh-Tu

AU - Lee, Ji-Young

AU - Ritter, Christian

AU - Thielemann, Roman

AU - Meyer, Janis

AU - Haselmann, Uta

AU - Funaya, Charlotta

AU - Laketa, Vibor

AU - Rohr, Karl

AU - Bartenschlager, Ralf

N1 - Copyright: © 2023 Pham et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

PY - 2023

Y1 - 2023

N2 - 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.

AB - 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.

KW - Humans

KW - Hepacivirus/physiology

KW - Lipopolysaccharides/metabolism

KW - Virus Assembly/physiology

KW - Hepatitis C

KW - Endosomes/metabolism

KW - Apolipoproteins E/metabolism

U2 - 10.1371/journal.ppat.1011052

DO - 10.1371/journal.ppat.1011052

M3 - Journal article

C2 - 37506130

VL - 19

JO - P L o S Pathogens (Online)

JF - P L o S Pathogens (Online)

SN - 1553-7374

IS - 7

M1 - e1011052

ER -

ID: 362102250