Loading…

Hepatitis C virus infection increases autophagosome stability by suppressing lysosomal fusion through an Arl8b-dependent mechanism

Autophagy is a conserved cellular process involving intracellular membrane trafficking and degradation. Pathogens, including hepatitis C virus (HCV), often exploit this process to promote their own survival. The aim of this study was to determine the mechanism by which HCV increases steady-state aut...

Full description

Saved in:
Bibliographic Details
Published in:The Journal of biological chemistry 2019-09, Vol.294 (39), p.14257-14266
Main Authors: Jones-Jamtgaard, Kellyann N., Wozniak, Ann L., Koga, Hiroshi, Ralston, Robert, Weinman, Steven A.
Format: Article
Language:English
Subjects:
Citations: Items that this one cites
Items that cite this one
Online Access:Get full text
Tags: Add Tag
No Tags, Be the first to tag this record!
cited_by cdi_FETCH-LOGICAL-c513t-269f86f2a863e7abe27f708b34367107f5c8b0abba39a2c910e388ac631390213
cites cdi_FETCH-LOGICAL-c513t-269f86f2a863e7abe27f708b34367107f5c8b0abba39a2c910e388ac631390213
container_end_page 14266
container_issue 39
container_start_page 14257
container_title The Journal of biological chemistry
container_volume 294
creator Jones-Jamtgaard, Kellyann N.
Wozniak, Ann L.
Koga, Hiroshi
Ralston, Robert
Weinman, Steven A.
description Autophagy is a conserved cellular process involving intracellular membrane trafficking and degradation. Pathogens, including hepatitis C virus (HCV), often exploit this process to promote their own survival. The aim of this study was to determine the mechanism by which HCV increases steady-state autophagosome numbers while simultaneously inhibiting flux through the autophagic pathway. Using the lysosomal inhibitor bafilomycin A1, we showed that HCV-induced alterations in autophagy result from a blockage of autophagosome degradation rather than an increase in autophagosome generation. In HCV-infected cells, lysosome function was normal, but a tandem RFP–GFP–LC3 failed to reach the lysosome even under conditions that activate autophagy. Autophagosomes and lysosomes isolated from HCV-infected cells were able to fuse with each other normally in vitro, suggesting that the cellular fusion defect resulted from trafficking rather than an inability of vesicles to fuse. Arl8b is an Arf-like GTPase that specifically localizes to lysosomes and plays a role in autophagic flux through its effect on lysosomal positioning. At basal levels, Arl8b was primarily found in a perinuclear localization and co-localized with LC3-positive autophagosomes. HCV infection increased the level of Arl8b 3-fold and redistributed Arl8b to a more diffuse, peripheral pattern that failed to co-localize with LC3. Knockdown of Arl8b in HCV-infected cells restored autophagosome–lysosome fusion and autophagic flux to levels seen in control cells. Thus, HCV suppresses autophagic flux and increases the steady-state levels of autophagosomes by increasing the expression of Arl8b, which repositions lysosomes and prevents their fusion with autophagosomes.
doi_str_mv 10.1074/jbc.RA119.008229
format article
fullrecord <record><control><sourceid>pubmed_cross</sourceid><recordid>TN_cdi_pubmedcentral_primary_oai_pubmedcentral_nih_gov_6768644</recordid><sourceformat>XML</sourceformat><sourcesystem>PC</sourcesystem><els_id>S0021925820349863</els_id><sourcerecordid>31383738</sourcerecordid><originalsourceid>FETCH-LOGICAL-c513t-269f86f2a863e7abe27f708b34367107f5c8b0abba39a2c910e388ac631390213</originalsourceid><addsrcrecordid>eNp1kUFr3DAQhUVpaLZp7z0V_QFvJMtryz0UliVtCoFCSSA3MZLHawVbMpK8sNf88mq7bWgP1WUE896TZj5CPnC25qyprp-0Wf_Yct6uGZNl2b4iK86kKMSGP74mK8ZKXrTlRl6StzE-sXyqlr8hl4ILKRohV-T5FmdINtlId_RgwxKpdT2aZL3LNxMQIkYKS_LzAHsf_YQ0JtB2tOlI9ZHGZZ4Dxmjdno7HeFLASPslnhLSEPyyHyg4ug2j1EWHM7oOXaITmgGcjdM7ctHDGPH973pFHr7c3O9ui7vvX7_ttneF2XCRirJue1n3JchaYAMay6ZvmNSiEnWTt9FvjNQMtAbRQmlazlBICabOw7Z5EeKKfD7nzouesDP5EwFGNQc7QTgqD1b923F2UHt_UHVTy7qqcgA7B5jgYwzYv3g5UyceKvNQv3ioM49s-fj3my-GPwCy4NNZgHnyg8WgorHoDHY2ZAqq8_b_6T8B1-2fTQ</addsrcrecordid><sourcetype>Open Access Repository</sourcetype><iscdi>true</iscdi><recordtype>article</recordtype></control><display><type>article</type><title>Hepatitis C virus infection increases autophagosome stability by suppressing lysosomal fusion through an Arl8b-dependent mechanism</title><source>BACON - Elsevier - GLOBAL_SCIENCEDIRECT-OPENACCESS</source><source>PubMed Central</source><creator>Jones-Jamtgaard, Kellyann N. ; Wozniak, Ann L. ; Koga, Hiroshi ; Ralston, Robert ; Weinman, Steven A.</creator><creatorcontrib>Jones-Jamtgaard, Kellyann N. ; Wozniak, Ann L. ; Koga, Hiroshi ; Ralston, Robert ; Weinman, Steven A.</creatorcontrib><description>Autophagy is a conserved cellular process involving intracellular membrane trafficking and degradation. Pathogens, including hepatitis C virus (HCV), often exploit this process to promote their own survival. The aim of this study was to determine the mechanism by which HCV increases steady-state autophagosome numbers while simultaneously inhibiting flux through the autophagic pathway. Using the lysosomal inhibitor bafilomycin A1, we showed that HCV-induced alterations in autophagy result from a blockage of autophagosome degradation rather than an increase in autophagosome generation. In HCV-infected cells, lysosome function was normal, but a tandem RFP–GFP–LC3 failed to reach the lysosome even under conditions that activate autophagy. Autophagosomes and lysosomes isolated from HCV-infected cells were able to fuse with each other normally in vitro, suggesting that the cellular fusion defect resulted from trafficking rather than an inability of vesicles to fuse. Arl8b is an Arf-like GTPase that specifically localizes to lysosomes and plays a role in autophagic flux through its effect on lysosomal positioning. At basal levels, Arl8b was primarily found in a perinuclear localization and co-localized with LC3-positive autophagosomes. HCV infection increased the level of Arl8b 3-fold and redistributed Arl8b to a more diffuse, peripheral pattern that failed to co-localize with LC3. Knockdown of Arl8b in HCV-infected cells restored autophagosome–lysosome fusion and autophagic flux to levels seen in control cells. Thus, HCV suppresses autophagic flux and increases the steady-state levels of autophagosomes by increasing the expression of Arl8b, which repositions lysosomes and prevents their fusion with autophagosomes.</description><identifier>ISSN: 0021-9258</identifier><identifier>EISSN: 1083-351X</identifier><identifier>DOI: 10.1074/jbc.RA119.008229</identifier><identifier>PMID: 31383738</identifier><language>eng</language><publisher>United States: Elsevier Inc</publisher><subject>ADP-Ribosylation Factors - genetics ; ADP-Ribosylation Factors - metabolism ; Arl8b ; Autophagosomes - metabolism ; autophagy ; Cell Biology ; Cell Line, Tumor ; GTPase ; Hepatitis C - metabolism ; Hepatitis C virus (HCV) ; Humans ; intracellular trafficking ; lysosome ; Lysosomes - metabolism ; Protein Transport</subject><ispartof>The Journal of biological chemistry, 2019-09, Vol.294 (39), p.14257-14266</ispartof><rights>2019 © 2019 Jones-Jamtgaard et al.</rights><rights>2019 Jones-Jamtgaard et al.</rights><rights>2019 Jones-Jamtgaard et al. 2019 Jones-Jamtgaard et al.</rights><lds50>peer_reviewed</lds50><oa>free_for_read</oa><woscitedreferencessubscribed>false</woscitedreferencessubscribed><citedby>FETCH-LOGICAL-c513t-269f86f2a863e7abe27f708b34367107f5c8b0abba39a2c910e388ac631390213</citedby><cites>FETCH-LOGICAL-c513t-269f86f2a863e7abe27f708b34367107f5c8b0abba39a2c910e388ac631390213</cites></display><links><openurl>$$Topenurl_article</openurl><openurlfulltext>$$Topenurlfull_article</openurlfulltext><thumbnail>$$Tsyndetics_thumb_exl</thumbnail><linktopdf>$$Uhttps://www.ncbi.nlm.nih.gov/pmc/articles/PMC6768644/pdf/$$EPDF$$P50$$Gpubmedcentral$$H</linktopdf><linktohtml>$$Uhttps://www.ncbi.nlm.nih.gov/pmc/articles/PMC6768644/$$EHTML$$P50$$Gpubmedcentral$$H</linktohtml><link.rule.ids>230,315,733,786,790,891,27957,27958,53827,53829</link.rule.ids><backlink>$$Uhttps://www.ncbi.nlm.nih.gov/pubmed/31383738$$D View this record in MEDLINE/PubMed$$Hfree_for_read</backlink></links><search><creatorcontrib>Jones-Jamtgaard, Kellyann N.</creatorcontrib><creatorcontrib>Wozniak, Ann L.</creatorcontrib><creatorcontrib>Koga, Hiroshi</creatorcontrib><creatorcontrib>Ralston, Robert</creatorcontrib><creatorcontrib>Weinman, Steven A.</creatorcontrib><title>Hepatitis C virus infection increases autophagosome stability by suppressing lysosomal fusion through an Arl8b-dependent mechanism</title><title>The Journal of biological chemistry</title><addtitle>J Biol Chem</addtitle><description>Autophagy is a conserved cellular process involving intracellular membrane trafficking and degradation. Pathogens, including hepatitis C virus (HCV), often exploit this process to promote their own survival. The aim of this study was to determine the mechanism by which HCV increases steady-state autophagosome numbers while simultaneously inhibiting flux through the autophagic pathway. Using the lysosomal inhibitor bafilomycin A1, we showed that HCV-induced alterations in autophagy result from a blockage of autophagosome degradation rather than an increase in autophagosome generation. In HCV-infected cells, lysosome function was normal, but a tandem RFP–GFP–LC3 failed to reach the lysosome even under conditions that activate autophagy. Autophagosomes and lysosomes isolated from HCV-infected cells were able to fuse with each other normally in vitro, suggesting that the cellular fusion defect resulted from trafficking rather than an inability of vesicles to fuse. Arl8b is an Arf-like GTPase that specifically localizes to lysosomes and plays a role in autophagic flux through its effect on lysosomal positioning. At basal levels, Arl8b was primarily found in a perinuclear localization and co-localized with LC3-positive autophagosomes. HCV infection increased the level of Arl8b 3-fold and redistributed Arl8b to a more diffuse, peripheral pattern that failed to co-localize with LC3. Knockdown of Arl8b in HCV-infected cells restored autophagosome–lysosome fusion and autophagic flux to levels seen in control cells. Thus, HCV suppresses autophagic flux and increases the steady-state levels of autophagosomes by increasing the expression of Arl8b, which repositions lysosomes and prevents their fusion with autophagosomes.</description><subject>ADP-Ribosylation Factors - genetics</subject><subject>ADP-Ribosylation Factors - metabolism</subject><subject>Arl8b</subject><subject>Autophagosomes - metabolism</subject><subject>autophagy</subject><subject>Cell Biology</subject><subject>Cell Line, Tumor</subject><subject>GTPase</subject><subject>Hepatitis C - metabolism</subject><subject>Hepatitis C virus (HCV)</subject><subject>Humans</subject><subject>intracellular trafficking</subject><subject>lysosome</subject><subject>Lysosomes - metabolism</subject><subject>Protein Transport</subject><issn>0021-9258</issn><issn>1083-351X</issn><fulltext>true</fulltext><rsrctype>article</rsrctype><creationdate>2019</creationdate><recordtype>article</recordtype><recordid>eNp1kUFr3DAQhUVpaLZp7z0V_QFvJMtryz0UliVtCoFCSSA3MZLHawVbMpK8sNf88mq7bWgP1WUE896TZj5CPnC25qyprp-0Wf_Yct6uGZNl2b4iK86kKMSGP74mK8ZKXrTlRl6StzE-sXyqlr8hl4ILKRohV-T5FmdINtlId_RgwxKpdT2aZL3LNxMQIkYKS_LzAHsf_YQ0JtB2tOlI9ZHGZZ4Dxmjdno7HeFLASPslnhLSEPyyHyg4ug2j1EWHM7oOXaITmgGcjdM7ctHDGPH973pFHr7c3O9ui7vvX7_ttneF2XCRirJue1n3JchaYAMay6ZvmNSiEnWTt9FvjNQMtAbRQmlazlBICabOw7Z5EeKKfD7nzouesDP5EwFGNQc7QTgqD1b923F2UHt_UHVTy7qqcgA7B5jgYwzYv3g5UyceKvNQv3ioM49s-fj3my-GPwCy4NNZgHnyg8WgorHoDHY2ZAqq8_b_6T8B1-2fTQ</recordid><startdate>20190927</startdate><enddate>20190927</enddate><creator>Jones-Jamtgaard, Kellyann N.</creator><creator>Wozniak, Ann L.</creator><creator>Koga, Hiroshi</creator><creator>Ralston, Robert</creator><creator>Weinman, Steven A.</creator><general>Elsevier Inc</general><general>American Society for Biochemistry and Molecular Biology</general><scope>6I.</scope><scope>AAFTH</scope><scope>CGR</scope><scope>CUY</scope><scope>CVF</scope><scope>ECM</scope><scope>EIF</scope><scope>NPM</scope><scope>AAYXX</scope><scope>CITATION</scope><scope>5PM</scope></search><sort><creationdate>20190927</creationdate><title>Hepatitis C virus infection increases autophagosome stability by suppressing lysosomal fusion through an Arl8b-dependent mechanism</title><author>Jones-Jamtgaard, Kellyann N. ; Wozniak, Ann L. ; Koga, Hiroshi ; Ralston, Robert ; Weinman, Steven A.</author></sort><facets><frbrtype>5</frbrtype><frbrgroupid>cdi_FETCH-LOGICAL-c513t-269f86f2a863e7abe27f708b34367107f5c8b0abba39a2c910e388ac631390213</frbrgroupid><rsrctype>articles</rsrctype><prefilter>articles</prefilter><language>eng</language><creationdate>2019</creationdate><topic>ADP-Ribosylation Factors - genetics</topic><topic>ADP-Ribosylation Factors - metabolism</topic><topic>Arl8b</topic><topic>Autophagosomes - metabolism</topic><topic>autophagy</topic><topic>Cell Biology</topic><topic>Cell Line, Tumor</topic><topic>GTPase</topic><topic>Hepatitis C - metabolism</topic><topic>Hepatitis C virus (HCV)</topic><topic>Humans</topic><topic>intracellular trafficking</topic><topic>lysosome</topic><topic>Lysosomes - metabolism</topic><topic>Protein Transport</topic><toplevel>peer_reviewed</toplevel><toplevel>online_resources</toplevel><creatorcontrib>Jones-Jamtgaard, Kellyann N.</creatorcontrib><creatorcontrib>Wozniak, Ann L.</creatorcontrib><creatorcontrib>Koga, Hiroshi</creatorcontrib><creatorcontrib>Ralston, Robert</creatorcontrib><creatorcontrib>Weinman, Steven A.</creatorcontrib><collection>ScienceDirect Open Access Titles</collection><collection>Elsevier:ScienceDirect:Open Access</collection><collection>Medline</collection><collection>MEDLINE</collection><collection>MEDLINE (Ovid)</collection><collection>MEDLINE</collection><collection>MEDLINE</collection><collection>PubMed</collection><collection>CrossRef</collection><collection>PubMed Central (Full Participant titles)</collection><jtitle>The Journal of biological chemistry</jtitle></facets><delivery><delcategory>Remote Search Resource</delcategory><fulltext>fulltext</fulltext></delivery><addata><au>Jones-Jamtgaard, Kellyann N.</au><au>Wozniak, Ann L.</au><au>Koga, Hiroshi</au><au>Ralston, Robert</au><au>Weinman, Steven A.</au><format>journal</format><genre>article</genre><ristype>JOUR</ristype><atitle>Hepatitis C virus infection increases autophagosome stability by suppressing lysosomal fusion through an Arl8b-dependent mechanism</atitle><jtitle>The Journal of biological chemistry</jtitle><addtitle>J Biol Chem</addtitle><date>2019-09-27</date><risdate>2019</risdate><volume>294</volume><issue>39</issue><spage>14257</spage><epage>14266</epage><pages>14257-14266</pages><issn>0021-9258</issn><eissn>1083-351X</eissn><notes>These authors contributed equally to this work.</notes><notes>Edited by George N. DeMartino</notes><abstract>Autophagy is a conserved cellular process involving intracellular membrane trafficking and degradation. Pathogens, including hepatitis C virus (HCV), often exploit this process to promote their own survival. The aim of this study was to determine the mechanism by which HCV increases steady-state autophagosome numbers while simultaneously inhibiting flux through the autophagic pathway. Using the lysosomal inhibitor bafilomycin A1, we showed that HCV-induced alterations in autophagy result from a blockage of autophagosome degradation rather than an increase in autophagosome generation. In HCV-infected cells, lysosome function was normal, but a tandem RFP–GFP–LC3 failed to reach the lysosome even under conditions that activate autophagy. Autophagosomes and lysosomes isolated from HCV-infected cells were able to fuse with each other normally in vitro, suggesting that the cellular fusion defect resulted from trafficking rather than an inability of vesicles to fuse. Arl8b is an Arf-like GTPase that specifically localizes to lysosomes and plays a role in autophagic flux through its effect on lysosomal positioning. At basal levels, Arl8b was primarily found in a perinuclear localization and co-localized with LC3-positive autophagosomes. HCV infection increased the level of Arl8b 3-fold and redistributed Arl8b to a more diffuse, peripheral pattern that failed to co-localize with LC3. Knockdown of Arl8b in HCV-infected cells restored autophagosome–lysosome fusion and autophagic flux to levels seen in control cells. Thus, HCV suppresses autophagic flux and increases the steady-state levels of autophagosomes by increasing the expression of Arl8b, which repositions lysosomes and prevents their fusion with autophagosomes.</abstract><cop>United States</cop><pub>Elsevier Inc</pub><pmid>31383738</pmid><doi>10.1074/jbc.RA119.008229</doi><tpages>10</tpages><oa>free_for_read</oa></addata></record>
fulltext fulltext
identifier ISSN: 0021-9258
ispartof The Journal of biological chemistry, 2019-09, Vol.294 (39), p.14257-14266
issn 0021-9258
1083-351X
language eng
recordid cdi_pubmedcentral_primary_oai_pubmedcentral_nih_gov_6768644
source BACON - Elsevier - GLOBAL_SCIENCEDIRECT-OPENACCESS; PubMed Central
subjects ADP-Ribosylation Factors - genetics
ADP-Ribosylation Factors - metabolism
Arl8b
Autophagosomes - metabolism
autophagy
Cell Biology
Cell Line, Tumor
GTPase
Hepatitis C - metabolism
Hepatitis C virus (HCV)
Humans
intracellular trafficking
lysosome
Lysosomes - metabolism
Protein Transport
title Hepatitis C virus infection increases autophagosome stability by suppressing lysosomal fusion through an Arl8b-dependent mechanism
url http://sfxeu10.hosted.exlibrisgroup.com/loughborough?ctx_ver=Z39.88-2004&ctx_enc=info:ofi/enc:UTF-8&ctx_tim=2024-09-21T21%3A22%3A23IST&url_ver=Z39.88-2004&url_ctx_fmt=infofi/fmt:kev:mtx:ctx&rfr_id=info:sid/primo.exlibrisgroup.com:primo3-Article-pubmed_cross&rft_val_fmt=info:ofi/fmt:kev:mtx:journal&rft.genre=article&rft.atitle=Hepatitis%20C%20virus%20infection%20increases%20autophagosome%20stability%20by%20suppressing%20lysosomal%20fusion%20through%20an%20Arl8b-dependent%20mechanism&rft.jtitle=The%20Journal%20of%20biological%20chemistry&rft.au=Jones-Jamtgaard,%20Kellyann%20N.&rft.date=2019-09-27&rft.volume=294&rft.issue=39&rft.spage=14257&rft.epage=14266&rft.pages=14257-14266&rft.issn=0021-9258&rft.eissn=1083-351X&rft_id=info:doi/10.1074/jbc.RA119.008229&rft_dat=%3Cpubmed_cross%3E31383738%3C/pubmed_cross%3E%3Cgrp_id%3Ecdi_FETCH-LOGICAL-c513t-269f86f2a863e7abe27f708b34367107f5c8b0abba39a2c910e388ac631390213%3C/grp_id%3E%3Coa%3E%3C/oa%3E%3Curl%3E%3C/url%3E&rft_id=info:oai/&rft_id=info:pmid/31383738&rfr_iscdi=true